US20180057791A1 - Methods and kits for cell activation - Google Patents

Methods and kits for cell activation Download PDF

Info

Publication number
US20180057791A1
US20180057791A1 US15/245,584 US201615245584A US2018057791A1 US 20180057791 A1 US20180057791 A1 US 20180057791A1 US 201615245584 A US201615245584 A US 201615245584A US 2018057791 A1 US2018057791 A1 US 2018057791A1
Authority
US
United States
Prior art keywords
cell
cells
nucleic acid
antibody
dna
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
US15/245,584
Other versions
US10294454B2 (en
Inventor
Ernest William Kovacs
Anup Sood
Reginald Donovan Smith
Evelina Roxana Loghin
Padmaparna Chadhuri
Vandana Keskar
Chrystal Mae Chadwick
Martin James Brown
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Global Life Sciences Solutions USA LLC
Original Assignee
General Electric Co
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by General Electric Co filed Critical General Electric Co
Assigned to GENERAL ELECTRIC COMPANY reassignment GENERAL ELECTRIC COMPANY ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CHADHURI, PADMAPARNA, KOVACS, ERNEST WILLIAM, BROWN, MARTIN JAMES, KESKAR, Vandana, SMITH, REGINALD DONOVAN, CHADWICK, CHRYSTAL MAE, SOOD, ANUP, LOGHIN, EVELINA ROXANA
Priority to US15/245,584 priority Critical patent/US10294454B2/en
Priority to CN201780051673.1A priority patent/CN109563484A/en
Priority to EP17769141.7A priority patent/EP3504323A1/en
Priority to PCT/US2017/048306 priority patent/WO2018039400A1/en
Priority to US16/339,077 priority patent/US11512288B2/en
Priority to JP2019510693A priority patent/JP7043114B2/en
Publication of US20180057791A1 publication Critical patent/US20180057791A1/en
Assigned to GENERAL ELECTRIC COMPANY reassignment GENERAL ELECTRIC COMPANY CORRECTIVE ASSIGNMENT TO CORRECT THE FIFTH ASSIGNOR CHADHURI, PADMAPARNA PREVIOUSLY RECORDED ON REEL 039525 FRAME 0388. ASSIGNOR(S) HEREBY CONFIRMS THE ASSIGNMENT. Assignors: CHAUDHURI, Padmaparna, KOVACS, ERNEST WILLIAM, BROWN, MARTIN JAMES, KESKAR, Vandana, SMITH, REGINALD DONOVAN, CHADWICK, CHRYSTAL MAE, SOOD, ANUP, LOGHIN, EVELINA ROXANA
Publication of US10294454B2 publication Critical patent/US10294454B2/en
Application granted granted Critical
Assigned to GLOBAL LIFE SCIENCES SOLUTIONS USA LLC reassignment GLOBAL LIFE SCIENCES SOLUTIONS USA LLC ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: GENERAL ELECTRIC COMPANY
Active legal-status Critical Current
Adjusted expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0636T lymphocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2809Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against the T-cell receptor (TcR)-CD3 complex
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2818Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against CD28 or CD152
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/50Cell markers; Cell surface determinants
    • C12N2501/51B7 molecules, e.g. CD80, CD86, CD28 (ligand), CD152 (ligand)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/50Cell markers; Cell surface determinants
    • C12N2501/515CD3, T-cell receptor complex

Definitions

  • T cell therapies in particular, have shown remarkable success in treating hematological tumors and show promise in the treatment of solid tumors. These treatments require isolation of a patient's PBMC's or T cells and subsequent expansion (activation and proliferation) to generate a personalized therapeutic dose.
  • this type of personalized therapy consists of removing blood cells from cancer patients; isolating and activating T cells; genetically modifying the T cells thereby programming those cells to recognize and attack cancer cells; expanding the T cells; and, lastly, introducing those cells back into the body so the patient's immune system can take over.
  • Activation is a critical component of the whole process as it is required for efficient introduction of genetic material and for robust expansion.
  • T cells comprising providing a population of T cells, adding a plurality of first agents, wherein the first agent comprises a T-cell activator and a first binder moiety; adding a second agent comprising a plurality of capture oligomers, wherein at least a segment of at least one of the plurality of capture oligomers is capable of associating with the first binder moiety; and incubating the population of T cells after steps (b) and (c), whereby at least a portion of the population of T cells is activated.
  • the method may further comprise the addition of a T-cell co-stimulator.
  • the present invention is a method for activating T cells, the method comprising providing a population of T cells, adding a plurality of first agents, wherein the first agent comprises a T-cell activator attached to a nucleic acid sequence; adding a nucleic acid polymer comprising a plurality of capture oligonucleotides, wherein at least a segment of at least one of the plurality of capture oligonucleotides is capable of associating with the nucleic acid sequence; and incubating the population of T cells after steps (b) and (c), whereby at least a portion of the population of T cells is activated.
  • the method may further comprise the addition of a T-cell co-stimulator.
  • the T-cell co-stimulator is attached to a nucleic acid sequence, wherein at least a segment of at least one of the plurality of capture oligonucleotides is capable of associating with the nucleic acid sequence attached to the T-cell co-stimulator.
  • the present invention relates to a kit comprising a T cell activator attached to a nucleic acid sequence; and a nucleic acid polymer comprising a plurality of capture oligonucleotide sequences, wherein the nucleic acid sequence is complementary to at least a segment of at least one of the capture oligonucleotide sequences.
  • FIG. 1A-1E are representative flow cytometry histograms for positively validated anti-CD3-o20b(+)act and anti-CD28-o20b(+)act conjugates binding to T cells.
  • 1 A Binding of a DNA (o20b(+)act) attached anti-CD28 antibody to T cells
  • 1 B Binding of corresponding unconjugated anti-CD28 antibody (positive control)
  • 1 C Binding of a DNA (o20b(+)act) attached anti-CD3 antibody to T cells
  • 1 D Binding of corresponding unconjugated anti-CD3 antibody (positive control)
  • 1 E Cell incubated with labeled secondary antibody (negative control).
  • FIG. 2 is an illustration of the preparation of single stranded rolling circle amplification (ssRCA) to produce RCA products (RCAact) for T cell activation in two steps.
  • ssRCA single stranded rolling circle amplification
  • FIG. 3A is graphical representation of T cell activation measured as population of cells expressing CD25 receptor (a marker of T cell activation) by DBTA (DNA-Based T cell Activation) relative to the CD3/CD28 Dynabeads benchmark with respect to both early (days 1 and 4) % CD25 expression. Also shown are various controls including unconjugated antibodies alone and cells alone (without activator) not expected to cause significant activation.
  • FIG. 3B is a graphical representation of the same cultures as in FIG. 3A at days 4 and 7 showing cell expansion represented as number of folds of expansion relative to starting count (cell counts).
  • FIG. 4A is a graphical representation showing significant activation (relative to control) achieved with different DBTA systems, a DBTA system wherein only the anti-CD3 antibody is attached to the o20b(+)act (anti-CD3-o20b(+)act) and another DBTA system wherein both anti-CD3 and anti-CD28 are attached to o20b(+)act. Measured is the % CD25 expression.
  • FIG. 4B is a graphical representation of the same cultures as FIG. 4A after 4 and 7 days showing x-fold expansion (cell counts) as in FIG. 3B .
  • FIG. 5A is a graphical representation showing a trend towards higher activation when the DBTA components are mixed in situ compared to when they are pre-incubated/pre-associated and then mixed with cells.
  • FIG. 5B is the same cultures as FIG. 5A after 4 days showing x-fold expansion (cell counts).
  • FIG. 6A is a graphical representation showing T cell activation (24-hour % CD25) under a variety of conditions including different ratios of anti-CD3-DNA and anti-CD28-DNA with same RCA amount (10 RCA repeat units/per anti-CD3-DNA conjugate) and different amounts of DBTA reagent while keeping the ratio of anti-CD3-DNA conjugate and RCA repeat unit constant at 1:10.
  • FIG. 6B is a graphical representation of cell expression of the same cultures as FIG. 6A after days 4 and 7 showing x-fold expansion (cell counts).
  • FIG. 7A is a graphical representation of greater than two-fold lower early activation (24-hour % CD25 expression) for sample where cells were pre-mixed with anti-CD3 and anti-CD28 DNA conjugates for 30 minutes prior to adding RCA compared to all three DTBA components added at once (one after another).
  • FIG. 7B is a graphical representation of the same cultures as FIG. 7A showing diminished day 7 cell expansion (x-fold expansion, cell counts).
  • FIG. 8A is a graphical representation showing T cell activation (% CD25 expression) is achieved for both CpG and no CpG RCA at different ratios (1:1, 1:10 and 1:100) of the anti-CD3-DNA:RCAact.
  • FIG. 8B is a graphical representation of the same cultures as FIG. 8A showing x-fold expansion (cell counts) after 4 and 7 days.
  • FIG. 9A is a graphical representation showing T cell activation with a different DNA sequence (a portion of MRSA (Methicillin-resistant Staphylococcus aureus ) sequence) attached to the anti-CD3 antibody and its complementary RCA product.
  • MRSA Metal-resistant Staphylococcus aureus
  • FIG. 9A is a graphical representation of the same cultures in FIG. 9A showing x-fold cell expansion after days 4 and 7.
  • FIG. 10A is a graphical representation showing that the larger the RCA product, the higher the activation (% CD25 expression).
  • FIG. 10B is a graphical representation of the same cultures in FIG. 10A showing higher x-fold expansion (cell counts) with larger product in early expansion phase (day 4), however, by day 7 all the three RCA products of different sizes yield comparable levels of cell expansion.
  • While certain embodiments are directed towards autologous cell therapies that involve collection, manipulation, and re-insertion of a patient's own cells, the applications of the disclosed techniques may include allogenic cells, modified human cells, or xenotransplantation of non-human cells.
  • Cell based therapies that are contemplated as being used in conjunction with the disclosed techniques may include therapies for organ or tissue regeneration, cancer treatment, blood disorders, immunotherapies, heart disease, or any other cell-based therapies.
  • a variety of cell types may be utilized within the context of the present invention including for example, but not limited to, cell types such as B cells, T-cells, or natural killer cells.
  • the cells can be isolated from any tissue such as peripheral blood, bone marrow, or tumor tissue.
  • Some embodiments are directed towards T cells enriched from peripheral blood by centrifugation using for example Ficoll-PaqueTM or PercollTM (GE Healthcare) gradient. Some other embodiments are directed towards a specific subpopulation of T-cells, such as CD28+, CD4+, CD8+, CD45RA+, and/or CD45RO+ T cells, further isolated by positive or negative selection techniques.
  • second agent refers to a polymer comprising a plurality of capture oligomers, wherein at least a segment of at least one of the plurality of capture oligomers is capable of associating with a first binder moiety.
  • the second agent is a nucleic acid polymer such as a rolling circle amplification product and the “capture oligomer” is a capture oligonucleotide sequence.
  • the nucleic acid polymer may comprise a plurality of the same or different capture oligonucleotide sequences.
  • the number of capture oligonucleotide sequences in the nucleic acid polymer is greater than 3, preferably greater than 30, and more preferably greater than 300.
  • the number of capture oligonucleotide sequences is greater than 2000.
  • the nucleic acid polymer may comprise a plurality of capture oligonucleotide sequences of the same or different nucleotide sequence, molecular weights, geometrical arrangements, and/or patterns of repetition.
  • the capture oligonucleotide sequence may have a length in range of from about 6 nucleotides to about 12 nucleotides, from about 12 nucleotides to about 25 nucleotides, from about 25 nucleotides to about 50 nucleotides, from about 50 nucleotides to about 100 nucleotides, from about 100 nucleotides to about 250 nucleotides, or from about 250 nucleotides to about 500 nucleotides.
  • the second agent is double-stranded nucleic acid polymer while in other embodiments it is a single-stranded nucleic acid polymer.
  • the nucleic acid polymer may include natural or unnatural nucleotides with modifications on the nucleic acid base, sugar or phosphate backbone.
  • the second agent may further comprise spacer oligonucleotide sequences having different number and/or sequence of nucleotides than the capture oligonucleotide sequences, or non-nucleic acid spacer molecules.
  • the second agent is a cationic polymer comprising a plurality of capture cationic oligomers, containing contiguous stretches of cationic monomers such as histidine or lysine.
  • the second agent may further comprise neutral or anionic spacer residues provided the overall charge remains cationic.
  • the second agent is an anionic polymer comprising a plurality of capture anionic oligomers, containing contiguous stretches of anionic monomers such as acrylate.
  • the second agent may further comprise neutral or cationic spacer residues provided the overall charge is anionic.
  • first agent refers to a cell activator attached to a “first binder moiety”.
  • the cell activator is a T-cell activator.
  • first binder moiety refers to a molecule that can associate with one or more capture oligomers.
  • the first binder moiety is a nucleic acid sequence and the “second agent” is a nucleic acid polymer comprising a plurality of capture oligonucleotide sequences, such that the nucleic acid sequence of the first binder moiety is complementary to at least a segment of at least one of the plurality of capture oligonucleotide sequences.
  • the nucleic acid sequence of the first binder moiety and the capture oligonucleotide sequence are capable of associating via complementary base-pair hybridization.
  • the first binder moiety and the capture oligonucleotide may hybridize to form a double or triple helix structure depending on whether the nucleic acid polymer is a double-stranded nucleic acid polymer or a single-stranded nucleic acid polymer.
  • all the nucleotide residues of at least one capture oligonucleotide sequence may hybridize to complementary nucleotides in the first binder moiety.
  • At least one capture oligonucleotide sequence may have 50 nucleotide residues and all the 50 nucleotide residues may hybridize to complementary nucleotides in the first binder moiety.
  • all the nucleotides of the capture oligonucleotide sequence may not have corresponding complementary nucleotides in the nucleic acid sequence of the first binder moiety.
  • the capture oligonucleotide sequence and/or the nucleic acid sequence of the first binder moiety may include nucleic acid analogs, with modified bases such as azidothymidine, inosine, or uridine, or modified sugars, e.g. 3′-OMe, or modified backbone, e.g. phosphorothioate, alkylphosphonate, phosphoramidate backbones.
  • the first or second agent may further comprise non-complementary spacer oligonucleotide sequences or non-nucleic acid spacer molecules.
  • the first binder moiety is an anionic moiety such as a nucleic acid sequence, alginate, polyglutamate, polyaspartate or hyaluronate and the second agent is a polymer comprising a plurality of cationic capture oligomers.
  • the anionic first binder moiety is capable of associating with at least a segment of at least one of the plurality of cationic capture oligomers via electrostatic association.
  • the first binder moiety is a cationic moiety and the second agent is a polymer comprising a plurality of anionic capture oligomers.
  • T-cell activator refers any agent that can activate a T-cell for proliferation and/or transduction.
  • suitable examples of T-cell activator include small organic molecules (for example ionomycin, phorbol myristate or acetate), natural or modified peptides, proteins (for example antibodies or affibodies), non-natural peptide mimics, nucleic acids (for example polynucleotides, PNA, DNA, RNA or aptamers), polysaccharides (for example lectins or sugars), lipids, enzymes, enzyme substrates or inhibitors, ligands, receptors, antigens, or haptens.
  • small organic molecules for example ionomycin, phorbol myristate or acetate
  • natural or modified peptides for example antibodies or affibodies
  • non-natural peptide mimics for example nucleic acids (for example polynucleotides, PNA, DNA, RNA or aptamers), polysaccharides (for
  • the T-cell activator binds to a T-cell surface receptor, such as a T-cell activating receptor, which results in delivery of a primary activation signal in a T-cell.
  • T-cell activating receptors include for example T-Cell Receptor (TCR) or CD3 receptor.
  • TCR T-Cell Receptor
  • a primary activation signal can be initiated through binding between a TCR and an antigen presented in conjunction with either MHC class I or class II molecules, in order to stimulate an antigen-specific T-cell activation.
  • a primary activation signal can also be initiated through binding between a CD3 receptor and a ligand targeted to the CD3 receptor, in order to stimulate a polyclonal T-cell activation.
  • the T-cell activator is an anti-CD3 antibody or fragments thereof.
  • T-cell activator include concanavalin A, protein kinase C (PKC) activator such as a phorbol ester (for example phorbol myristate acetate) or calcium ionophore (for example ionomycin which raises cytoplasmic calcium concentrations) and others.
  • PLC protein kinase C
  • a first agent that binds to a TCR or a CD3 receptor may be used in conjunction with other T-cell activators.
  • the method of activating T cells comprises the addition of a plurality of a 1 st population of first agents, wherein all the first agents in the 1 st population of first agents comprises T-cell activator of the same type, such as an anti-CD3 antibody.
  • the method may further comprise the addition of a 2 nd , 3 rd , . . . n th population of first agents.
  • the T-cell activators among different populations of first agents may be of the same type or different types such as anti-CD3 antibodies or fragments thereof or antigens bound to MHC molecules.
  • the method of activating T cells further comprises the addition of a T-cell co-stimulator.
  • T-cell co-stimulators include ligands targeted towards T-cell co-stimulatory receptors such as CD28, CD2, ICOS, OX40, or 4-IBB receptors.
  • Suitable ligands may include one or more of natural or modified peptides, proteins (for example antibodies, affibodies), non-natural peptide mimics, nucleic acids (for example polynucleotides, PNA, DNA, RNA, or aptamers), polysaccharides (for example lectins, sugars), lipids, enzymes, enzyme substrates or inhibitors, ligands, receptors, antigens, or haptens.
  • the addition of a T-cell co-stimulator may include the addition of one or more types of T-cell co-stimulators. In some embodiments the T-cell co-stimulator may be attached to a second binder moiety.
  • second binder moiety refers to a molecule that can associate with one or more capture oligomers.
  • the second agent is a nucleic acid polymer and the second binder moiety is a nucleic acid sequence, which is same as or different from the nucleic acid sequence of the first binder moiety.
  • the second agent is a cationic polymer and the second binder moiety is an anionic moiety.
  • the second agent is an anionic polymer and the second binder moiety is a cationic moiety.
  • a T-cell activator can be attached to a first binder moiety by any means provided the attachment does not interfere with the activator's ability to activate T cells.
  • a T-cell co-stimulator can be attached to a second binder moiety by any means provided the attachment does not interfere with the co-stimulator's ability to provide a co-stimulatory signal to the T cell.
  • the attachment may be covalent or noncovalent, electrostatic, or hydrophobic. In a preferred embodiment the attachment is covalent.
  • the T-cell activator is an anti-CD3 antibody attached to a nucleic acid sequence
  • the T-cell co-stimulator is an anti-CD28 antibody attached to a nucleic acid sequence
  • the second agent is a rolling circle amplification product comprising a plurality of complementary capture oligonucleotide sequences.
  • the first agent and the second agent are soluble in cell culture media.
  • “cell culture media” refers to any standard T cell culture media, which are known in the art. Illustrative media include but are not limited to RPMI 1640, AIM-V, DMEM, MEM, ⁇ -MEM, F-12, X-Vivo 15.
  • the T cell culture media may be serum-free or supplemented with serum, such as human serum or serum supplement.
  • the T cell culture media is further supplemented with additional growth factors and cytokines such as interleukin-2 (Il-2), interleukin-7 (Il-7) or interleukin-15 (IL-15).
  • situ generally refers to an event occurring in the original location, for example in the location where T cell activation occurs. In some embodiments “in situ” refers to an event happening in the T cell culture media under cell culturing condition.
  • the invention includes embodiments that relate generally to methods of activating T cells for cellular therapy.
  • the method includes the addition of a plurality of first agents, each first agent comprising a T-cell activator attached to a first binder moiety, wherein the T-cell activators are capable of binding to a set of T-cell surface receptors, such as the T-cell activating receptors, inducing a cellular event within the T cells that leads to activation and proliferation of the cells.
  • a clustering of the T-cell surface receptors is induced by a second agent, which associates with the first binder moiety bringing the T-cell activators in proximity to each other.
  • the method further includes the addition of a plurality of T-cell co-stimulators capable of binding to a different set of T-cell surface receptors, such as the T-cell co-stimulatory receptors.
  • the T-cell co-stimulator may be attached to a second binder moiety.
  • the second agent associates with both the first binder moiety and the second binder moiety thereby inducing a clustering of the T-cell surface receptors.
  • the second agent is a nucleic acid polymer comprising a plurality of capture oligonucleotide sequences and the association is caused by base-pair hybridization between a nucleic acid sequence attached to the T-cell activator and a complementary capture oligonucleotide sequence.
  • the method further comprises the addition of a T-cell co-stimulator attached to a second binder moiety, wherein the second binder moiety is also a nucleic acid sequence, and the nucleic acid sequence is capable of association with a complementary capture oligonucleotide sequence by base-pair hybridization.
  • the association of the capture oligomer with the first binder moiety and the second binder moiety is caused by electrostatic interaction between oppositely charged species for example anionic moieties attached to the T-cell activator and T-cell co-stimulator with cationic capture oligomers or vice versa.
  • the first agent, the second agent, and optionally a T-cell co-stimulator which may or may not be attached to a second binder moiety, are added simultaneously or sequentially in any order, to a population of T cells provided in a cell culture media, to allow an in-situ association.
  • the first agent, the second agent, and optionally a T-cell co-stimulator attached to a second binder moiety are allowed to pre-incubate and associate to form a pre-associated complex. The pre-associated complex is then added to the population of T cells.
  • the first agent, the second agent, and optionally the T-cell co-stimulator attached to a second binder moiety are allowed to associate in-situ.
  • the T cells are incubated with the first agent, the second agent, and optionally a T-cell co-stimulator for a period of time, whereby at least a portion of the population of T cells is activated.
  • the T-cell co-stimulator is attached to a second binder moiety.
  • the time sufficient to activate a portion of the population of T cells may range from about 1 minute to about 14 days. In certain embodiments, the time may be from about 24 h to about 8 days. In some embodiments, at least 15% of T cells are activated. In preferred embodiments at least 25% of the T cells are activated, and in more preferred embodiment the majority of the T cells, greater than 50% are activated.
  • the incubation may be done in a bioreactor having a controlled environment of temperature, humidity, CO2 concentration, for example at 37° C. and 5% CO2. In some embodiments the incubation may be done in a static bioreactor where there is no movement of the bioreactor, while in some other embodiments the incubation may be done in a bioreactor placed on a rocking platform.
  • the invention provides methods to activate and selectively expand a specific subpopulation of T cells from a mixed population of T cells. This can be achieved for example by varying the nature or relative proportion of the T-cell activators and T-cell co-stimulators. Further, the invention provides methods to control T-cell surface receptor clustering and hence activation by adjusting for example the number of or distance among the T-cell activators and T-cell co-stimulators, which are associated with the second agent. In an exemplary embodiment where the second agent is a nucleic acid polymer, this can be achieved by precisely controlling the number and/or length of the capture oligonucleotides, or the length of the spacers.
  • the expression of certain antibodies such as CD25 may be used to measure the activation of the T cells.
  • the expression of CD25 receptors on the T cells may be assayed by labeled anti-CD25 antibodies to enumerate the labeled cells.
  • activation may be measured by other markers signifying T-cell surface receptor clustering, which induces activation and expansion.
  • the other markers used to measure T cell activation and expansion include CD4, CD8, CD27, CD28, CD3, CD57, CD25, and CD62L.
  • the first agent and the second agents are bio-degradable.
  • bio-degradable refers to materials that degrade in biological fluid. In some embodiments, the degradation may occur using chemical or enzymatic means.
  • the method of activation of T cells further comprises the addition of a degrading enzyme such as a nuclease. In some embodiments the degrading enzyme, such as the nuclease, is added after at least a portion of the population of T cells is activated. In some embodiments, the degrading enzyme is added after the T cells have expanded at least 10-100 fold.
  • the degrading enzyme is added at the end of the culture period, before the T cells are harvested from the cell culture media, prior to washing, concentration and final formulation. It should be noted that the degrading enzyme can be added any time after at least a portion of the population of T cells is activated and before the T cells are administered into a patient. In certain embodiments, after degradation, the degraded by-products of the first and second agents can be removed during washing and concentration of the T cells, and additional purification steps are not necessary. In some other embodiments, the second agent and the first agent are biocompatible and may be rapidly degraded in the blood stream. In such cases, the addition of a degrading enzyme may not be required before the T cells are administered into a patient.
  • the use of such soluble, biodegradable systems are advantageous over polystyrene beads or other comparable bead based approaches since the use of such systems may avoid the need for additional purification steps, such as magnetic separation, which often leads to significant cell loss.
  • the second binder moiety is also biodegradable.
  • the method of T cell activation further comprises the addition of a vector comprising a foreign gene.
  • the foreign gene encodes a chimeric antigen receptor or a T-cell receptor.
  • the vector is a viral vector such as a ⁇ -retroviral vector or a lentiviral vector.
  • the vector is a plasmid vector.
  • the vector is added simultaneously with the first agent, while in some other embodiments, the vector is added after at least a portion of the population of T cells is activated.
  • kits comprising a T-cell activator attached to a nucleic acid sequence, a nucleic acid polymer comprising a plurality of capture oligonucleotide sequences, and optionally a T-cell co-stimulator.
  • the T-cell co-stimulator is attached to a nucleic acid sequence, which is same as or different from the nucleic acid sequence attached to the T-cell activator.
  • the kit is a DNA-Based T cell Activation (DBTA) construct comprising a DNA polymer such as a rolling circle amplification product and one or more of the following components, i) a T-cell activator attached to a DNA sequence (first binder moiety), and ii) a T-cell co-stimulator attached to a DNA sequence (second binder moiety), which is same as or different from the DNA sequence attached to the T-cell activator.
  • DBTA DNA-Based T cell Activation
  • DNA-Based T cell Activation (DBTA) polymer constructs are representative nucleic acid polymer systems that initiate T cell clustering, activation, and subsequent expansion.
  • DBTA comprises a DNA polymer produced via rolling circle amplification (such as RCAact) and consisting of repeating 43-base oligonucleotide segments and one or more of the following components: (1) anti-human CD3 monoclonal antibodies conjugated to 20-base DNA sequence derived from human beta-actin (o20b(+)act), (2) anti-human CD28 monoclonal antibodies also conjugated with DNA sequence o20b(+)act.
  • the 43-base oligonucleotide segments bear full complementarity to the DNA sequences conjugated to the antibodies (1) and (2).
  • the sequences of RCAact are derived from the complement to the RCA template sequences given below in Table 1:
  • the starting nucleic acid sequence (prot-mal-o20b(+)act) is provided by TriLink Biotechnologies and comprises a 20-base DNA sequence capped with an N-terminal C6 spacer followed by a protected tricyclic maleimide moiety.
  • a reactive mal-o20b(+)act derivative is generated via an inverse electron-demand Diels-Alder deprotection step, whereby the starting nucleic acid sequence is suspended in anhydrous toluene ( ⁇ 1 mg/mL) and heated at 90° C. for 4 h. Precipitation of the nucleic acid sequences via benchtop centrifugation and removal of solvent is followed by several washes (3 ⁇ 1 mL) with cold ethanol.
  • the washed solid product Upon further reduction of residual organic solvents under reduced pressure, the washed solid product is dissolved in 100 mM HEPES buffer, pH 7.3 and the solution concentration is determined via UV-Vis spectroscopy (NanoDrop).
  • the resulting stock solution (0.5-1 mM in DNA) or a portion thereof may be used directly for antibody conjugation with thiol-modified antibody.
  • the remaining mal-o20b(+)act stock may be stored for several months at ⁇ 20° C. without significant loss of reactivity.
  • a 10 mM stock solution of Traut's reagent (2-aminothiolane hydrochloride) is first prepared in 100 mM HEPES buffer, pH 7.3.
  • a 1 mg/mL solution of anti-CD3 Ab (OKT3 clone, eBioscience) or anti-CD28 Ab (9.3 clone, GeneTex) in PBS is then mixed with both 20 ⁇ pH 8.5 borate buffer (ThermoFisher) and 10 mM Traut's reagent stock solution in a 8:1:1 ratio by volume.
  • the resulting solution is thoroughly mixed and allowed to incubate at room temperature for 0.75 h.
  • the unused portion of the Traut's mixture may be stored for several months at ⁇ 20° C.
  • the reaction mixture is purified using a conventional desalting column (e.g. NAP-5 or PD-10, GE Healthcare Life Sciences) that has been equilibrated with 100 mM HEPES, pH 7.3 buffer.
  • the collected fractions are then immediately analyzed by UV-Vis spectroscopy (NanoDrop).
  • the resulting protein recovery at this stage is typically >60% for both anti-CD3 and anti-CD28 antibodies using known molar extinction coefficients for antibody at 280 nm.
  • the volume of mal-o20b(+)act corresponding to a target molar input ratio of 10-40:1 o20b(+)act:Ab is added to an aliquot of freshly prepared, purified, thiol-activated Ab. After thorough mixing, the resulting solution is allowed to incubate at room temperature overnight (16-24 h).
  • Final conjugate purification is achieved via selective precipitation of Ab using a saturated ammonium chloride solution. First, a volume of saturated ammonium chloride equal to the total reaction volume is added, thoroughly mixed, and placed on ice. After 15 min, the sample is centrifuged at 15,000 ⁇ g rcf for 10 min at 10° C.
  • Example 1 To ensure that the Ab-DNA conjugates prepared in Example 1 retain their specific cell binding capabilities to T cells, validation studies are performed for each conjugate batch using flow cytometry (Cytoflex S, Beckman Coulter). NBP Pan T Cells (ALL Cells) are thawed at 37° C. in 10 mL warm complete media (see Example 4) and then centrifuged at 300 ⁇ g rcf for 10 min. Cells are then resuspended in 10 mL fresh complete media and analyzed on a Nucleocounter® NC-200 system to determine cell counts and viability. After adjusting the concentration to 1 ⁇ 10 6 cells/mL and washing with PBS, the T cells are then blocked in 10% Normal Goat Serum (NGS) in PBS at 4° C.
  • NGS Normal Goat Serum
  • anti-CD3-DNA and/or anti-CD28-DNA conjugate (anti-CD3-o20b(+)act and/or anti-CD28-o20b(+)act) test samples (Example 1) as well as unlabeled or unconjugated anti-CD3 and/or anti-CD28 Ab positive controls (Ab not attached to DNA) are used.
  • FIG. 1A through 1E Representative flow cytometry histograms for positively validated anti-CD3-o20b(+)act and anti-CD28-o20b(+)act conjugates binding to T cells are shown in FIG. 1A through 1E ; Anti-CD28-o20b(+)act, anti-CD28 unlabeled, Anti-CD3o20b(+)act, anti-CD3 unlabeled, and FITC-labeled respectively.
  • high percentage of anti-CD3+ and anti-CD28+ cell binding is observed (>85% for both).
  • Preparation of single stranded rolling circle amplification (ssRCA) to produce RCA products (RCAact) for T cell activation comprises two steps: 1) ligation of a linear DNA strand to generate a circular template, and 2) amplification of the ligated circle to synthesize long single stranded concatemers.
  • the ligation was accomplished using a high concentration of T4 DNA Ligase and its corresponding reaction buffer (New England Biolabs®) Two deoxyoligonucleotides (oligonucleotide) were present in the ligation.
  • oligonucleotide oligonucleotide
  • One oligonucleotide (RCAact-NoCG) consisted of 43 bases of the DNA sequence of the human ⁇ -actin gene and contained a 5′ phosphate group.
  • This 43 base oligonucleotide would become the circular template following ligation.
  • the second oligonucleotide was 20 bases in length (RCAact Primer) and was complimentary to both ends of the 43 base oligonucleotide. This second oligonucleotide was used to both form the circle prior to ligation and then amplify the circle in the subsequent ssRCA reaction (see FIG. 2 ).
  • 450 pmol of the 43 base oligonucleotide were mixed in a 120 ⁇ L volume with 300 pmol of the 20 base oligonucleotide in an annealing buffer consisting of 10 mM Tris, pH 8, and 50 mM sodium chloride. The mixture was heated at 95° C.
  • the ssRCA reaction was prepared by mixing 69.3 ⁇ L of the completed ligation reaction with 550 ⁇ L of 2 ⁇ Phi29 Reaction Buffer (100 mM HEPES Buffer, pH 8.0, 150 mM potassium chloride, 2 mM TCEP, 40 mM magnesium chloride, 0.02% (v/v) Tween 20, 5% (v/v) polyethylene glycol and 1.6 mM each dATP, dCTP, dGTP and dTTP) in a final volume of 1.078 mL. After mixing, amplification was started by the addition of 22 ⁇ L of 1 mg/ml Phi29 DNA polymerase. Amplification reactions were incubated at 30° C. for 18 hours and then 65° C.
  • Phi29 Reaction Buffer 100 mM HEPES Buffer, pH 8.0, 150 mM potassium chloride, 2 mM TCEP, 40 mM magnesium chloride, 0.02% (v/v) Tween 20, 5% (
  • RCAact products The apparent size of RCAact products is determined by pulse field gel electrophoresis relative to yeast ( S. cerevisiae ) or lambda DNA molecular weight ladders.
  • gel-shift assays using non-denaturing agarose gels are used to confirm hybridization of anti-CD3-o20b(+)act and anti-CD28-o20b(+)act conjugates (Example 1) with RCAact.
  • Example 4 Comparison of T Cell Activation and Expansion Using Dynabeads® Human T-Expander CD3/CD28 and a DBTA System of Nucleic Acid Polymers with Ab-DNA Conjugates (Anti-CD3-DNA and Anti-CD28-DNA)
  • Pan T Cells Frozen aliquots of human Pan T Cells from AllCells (Catalog# PB009-IF) are used for all activation and expansion studies. Pan T Cells are thawed and processed as described in Example 2 and added to following complete X-Vivo media to give an initial concentration of 1 ⁇ 106 cells/mL as shown in Table 2.
  • Typical activation and expansion experiments are performed in a 6-well format using a 2 mL seeding volume per well with a minimum of one additional replicate per condition tested.
  • Dynabeads® Human T-Expander CD3/CD28 (Catalog#111.41D, ThermoFisher) are prepared according to the manufacturer's instructions. Briefly, a bead aliquot is washed with 0.3 mL complete media three times, with supernatant wash removal occurring after sample application to DynaMag-2 permanent magnet (ThermoFisher). After resuspension in complete media, a 60 bead slurry aliquot is added per well such that an approximate 3:1 bead-to-cell ratio is used for initial activation conditions (starting with 2 ⁇ 10 6 total cells per well).
  • DBTA components Ab-DNA and RCAact
  • aliquots of DBTA components are added separately and consecutively to cells directly from their respective stock solutions (stored at 4° C.). Additions of each DBTA component may be made in any sequential order.
  • Standard initial concentrations of DBTA components are as follows: 1 ⁇ g/mL (6.7 nM) each for anti-CD3-o20b(+)act (anti-CD3-DNA) and anti-CD28-o20b(+)act (anti-CD28-DNA) with 10-fold molar excess of RCAact ( ⁇ 67 nM).
  • RCAact concentration is based on the molar concentration of the repeating 43-base segment and independent of total RCA product length or polydispersity.
  • the well contents are mixed with a 1 mL pipette.
  • the plates are then incubated at 37° C. and 5% CO2 atmosphere under static conditions for 1-7 days or longer with periodic aliquots taken and, dilutions done with fresh media as needed for continuing expansion and analysis. Confirmation of cell activation is achieved by % CD25 expression using flow cytometry after 24 h incubation (day 1) and followed by additional measurements, if needed, at days 4 and 7. Cell counts, viability, and size (blasting) are additionally measured at days 4 and 7 using the Nucleocounter.
  • a 1:4 or 1:8 dilutions with fresh complete media is performed to reduce cell density to 250,000 cells/mL per well. This enables cells to expand in the exponential phase, without overgrowth, until day 7 analysis.
  • cell density is maintained at a minimum of 500,000 cells/mL. Further phenotypic analysis via flow cytometry may be conducted at day 7 for select samples of interest.
  • the panel of cell surface markers under investigation includes CD4, CD8, CD27, CD28, CD3, CD57, CD25, and CD62L.
  • FIGS. 3A and 3B shows clear, comparable performance of DBTA relative to the CD3/CD28 Dynabeads benchmark with respect to both early (days 1 and 4) % CD25 expression as well as x-fold cell expansion after days 4 and 7.
  • Control samples with unmodified antibodies, RCA alone, unmodified antibodies with unconjugated nucleic acid sequence (free oligo) and cells only show very low level of activation and expansion, as expected.
  • the robustness of the DBTA system is also confirmed as the standard deviations of activation and expansion match those observed with Dynabeads.
  • FIGS. 3A and 3B includes data from five separate experiments, seven individual human T cell donors, and 4 different batches of antibody conjugates.
  • FIG. 4A and 4B show that significant activation and expansion (relative to control) can also be achieved with a DBTA system wherein only the anti-CD3 antibody is attached to the o20b(+)act (anti-CD3-o20b(+)act).
  • FIG. 4A shows the CD25 expression of the cultures after 24 hrs while FIG. 4B shows the CD25 expression of the same cultures after 4 and 7 days of expansion. However, the best performance is achieved when both anti-CD3 and anti-CD28 antibodies are attached to DNA to form the Ab-DNA conjugates.
  • the data for FIG. 4 represents averages taken from over 5 separate experiments encompassing over six individual human T cell donors.
  • Example 5 Comparison of Large-Scale T Cell Activation and Expansion Using Dynabeads® (ThermoFisher Scientific, Waltham, Mass.) Human T-Expander CD3/CD28 and a DBTA System of Nucleic Acid Polymers (RCAact) with Ab-DNA Conjugates (Anti-CD3-o20b(+)Act and Anti-CD28-o20b(+)Act)
  • Example 4 In addition to 6-well plate studies, a large scale comparison of T cell activation and expansion efficiency using 72C VueLife® bags (Cell Genix GmbH, Breisgau, Germany) was also undertaken. The same ratios and components described in Example 4 were scaled linearly to accommodate starting conditions of 32 ⁇ 10 6 cells in 32 mL of complete X-Vivo media (1 ⁇ 106 cells/ml). The cultures were maintained in the 72C VueLife bags within a standard cell culture incubator. On Day 4, cells were collected, washed with fresh media, counted, diluted in appropriate media volume to give 0.5 ⁇ 106 cells/mL and re-seeded within the VueLife 72C.
  • FIGS. 5A and 5B shows that comparable levels of both early activation (24-hour % CD25 expression) and day 4 cell expansion is achieved whether or not the all the DBTA components are pre-incubated/pre-associated (anti-CD3-DNA/anti-CD28-DNA DBTA pre-associated) together or added simultaneously at the start of cell activation (anti-CD3-DNA/anti-CD28-DNA DBTA in-situ)
  • FIG. 5A is after 24 hr incubation and 5 B is after 4 days. Both sets of DBTA samples likewise compare favorably to benchmark Dynabead numbers for activation and early expansion. This data is derived from two separate experiments featuring four different human T cell donors and four different batches of Ab-DNA conjugates.
  • FIG. 6A shows that sufficient T cell activation (24-hour % CD25 expression) and FIG. 6B cell expansion (days 4 and 7) are achieved at a variety of input quantities of antibody and RCAact product.
  • the lowest input quantities of both antibody conjugate (0.25 ⁇ g/mL input or ⁇ 170 pM) and the corresponding RCAact product ( ⁇ 17 nM) showed the lowest levels of 24-hour % CD25 expression (60%), although this level of activation still far exceeds the cells-only negative control (20%) and eventually leads to day 7-fold expansion that matches the benchmark Dynabead samples.
  • FIG. 7A highlights a greater than two-fold lower early activation (24-hour % CD25 expression) and FIG. 7B shows diminished day 7 cell expansion for the Ab-DNA/cell pre-incubated sample (anti-CD3-DNA/anti-CD28-DNA DBTA (Ab-DNA/cells pre-incubated)) relative to both standard DBTA samples (featuring simultaneous addition of all components) (anti-CD3-DNA/anti-CD28-DNA DBTA in situ) and Dynabead controls.
  • FIG. 7B also shows results of expansion after 4 days.
  • FIGS. 8A and 8B shows that optimal T cell activation (% CD25 expression) is achieved for both CpG and no CpG RCA at the anti-CD3-DNA:RCAact ratio of 1:10. Diminished activation efficiency is observed at both higher and lower ratios of Ab-DNA-to-RCA. All samples, however, demonstrate day 7 T cell expansion of about 10-fold or greater, matching or exceeding the Dynabead control sample.
  • FIG. 8A is after 1 and 4 day culture showing % CD25
  • FIG. 8B is after 4 and 7 days showing x-fold expansion.
  • Example 10 Demonstration of Human T Cell Activation and Expansion Using an Alternative DBTA System with a Different Sequence and Oligo Length
  • Sequence (5′ ⁇ length 3′) including SEQ ID Name sense (b) modifications NO use/application RCA primer - 20 ATC AAT GAT 5 primer for RCA reactions, sequence derived (MRSA) GCA TAA CAT CT from MRSA genome RCA template + 43 /Phos/CAT CAT 6 template for RCA reactions, sequence derived (MRSA) TGA TTT AGA from MRSA genome CAC TGA AAA AGT TCG AGG AGA TGT TAT G o25b(+)mrsa + 25 /MalC6/CAT CAT 7 conjugation to Ab via 5'-maleimide and binding TGA TTT AGA to MRSA-derived RCA product CAC TGA AAA A
  • FIGS. 9 A and 9 B shows clear, comparable performance of DBTA component (anti-CD3-DNA conjugate only with soluble anti-CD28 Ab) relative to the CD3/CD28 Dynabeads benchmark with respect to both early (days 1 and 4) % CD25 expression ( FIG. 9A ) as well as x-fold cell expansion after days 4 and 7 ( FIG. 9B ).
  • DBTA component anti-CD3-DNA conjugate only with soluble anti-CD28 Ab
  • Example 11 Demonstration of Human T Cell Activation and Expansion Using a DBTA System Featuring Cleaved RCA Polymeric Products
  • FIGS. 10A and 10B shows that the larger the RCA product, the higher the activation ( FIG. 10A % CD25 expression) and fold-expansion for the early days 1-4 of the experiment, while the trend becomes less pronounced later in the study. In particular, by day 7, all the three RCA products of different sizes yield comparable levels of cell expansion ( FIG. 10B ).

Landscapes

  • Health & Medical Sciences (AREA)
  • Immunology (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Genetics & Genomics (AREA)
  • General Health & Medical Sciences (AREA)
  • Biochemistry (AREA)
  • Biomedical Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Zoology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biophysics (AREA)
  • Wood Science & Technology (AREA)
  • Biotechnology (AREA)
  • Hematology (AREA)
  • Cell Biology (AREA)
  • General Engineering & Computer Science (AREA)
  • Microbiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Medicinal Preparation (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

Provided are methods and kits for activating T cells, the method comprising providing a population of T cells, adding a plurality of first agents, where the first agent comprises a T-cell activator and a first binder moiety, and adding a second agent comprising a plurality of capture oligomers, where at least a segment of at least one of the plurality of capture oligomers is capable of associating with the first binder moiety. The method further comprises incubating the population of T cells, whereby at least a portion of the population of T cells is activated.

Description

    SEQUENCE LISTING
  • The instant application contains a Sequence Listing which has been submitted electronically in ASCII format and is hereby incorporated by reference in its entirety. Said ASCII copy, created on Sep. 26, 2016, is named 312704-1_SL.txt and is 2,065 bytes in size.
  • BACKGROUND
  • Cellular therapies, T cell therapies in particular, have shown remarkable success in treating hematological tumors and show promise in the treatment of solid tumors. These treatments require isolation of a patient's PBMC's or T cells and subsequent expansion (activation and proliferation) to generate a personalized therapeutic dose.
  • For the most part this type of personalized therapy consists of removing blood cells from cancer patients; isolating and activating T cells; genetically modifying the T cells thereby programming those cells to recognize and attack cancer cells; expanding the T cells; and, lastly, introducing those cells back into the body so the patient's immune system can take over. Activation is a critical component of the whole process as it is required for efficient introduction of genetic material and for robust expansion.
  • Several technology platforms exist in the commercial space, super paramagnetic, nonpyrogenic polystyrene beads with antibodies covalently bound to the surface, such as Dynabeads® CD3/CD28 CTS' (Life Technologies, Beverly, Mass.) is one of the most widely used to provide for isolation, activation and expansion of T cells. Still, in using bead based clustering and cell activation, significant cell loss is observed using the bead platform. This is related to bead removal after cell expansion is complete. Alternate technologies, such as the activator from StemCell Technologies that circumvent the issue of bead removal, are not very effective for cell activation, particularly early in the activation phase when the viral transduction is generally carried out.
  • As such a new technology platform is desired that will provide novel approaches to cluster the T-cell surface receptors required for T cell activation and provide co-stimulatory signals to increase proliferation.
  • BRIEF DESCRIPTION
  • Disclosed herein are methods for activating T cells, the method comprising providing a population of T cells, adding a plurality of first agents, wherein the first agent comprises a T-cell activator and a first binder moiety; adding a second agent comprising a plurality of capture oligomers, wherein at least a segment of at least one of the plurality of capture oligomers is capable of associating with the first binder moiety; and incubating the population of T cells after steps (b) and (c), whereby at least a portion of the population of T cells is activated. The method may further comprise the addition of a T-cell co-stimulator.
  • In some embodiments, the present invention is a method for activating T cells, the method comprising providing a population of T cells, adding a plurality of first agents, wherein the first agent comprises a T-cell activator attached to a nucleic acid sequence; adding a nucleic acid polymer comprising a plurality of capture oligonucleotides, wherein at least a segment of at least one of the plurality of capture oligonucleotides is capable of associating with the nucleic acid sequence; and incubating the population of T cells after steps (b) and (c), whereby at least a portion of the population of T cells is activated. The method may further comprise the addition of a T-cell co-stimulator. In some embodiments the T-cell co-stimulator is attached to a nucleic acid sequence, wherein at least a segment of at least one of the plurality of capture oligonucleotides is capable of associating with the nucleic acid sequence attached to the T-cell co-stimulator.
  • In some embodiments the present invention relates to a kit comprising a T cell activator attached to a nucleic acid sequence; and a nucleic acid polymer comprising a plurality of capture oligonucleotide sequences, wherein the nucleic acid sequence is complementary to at least a segment of at least one of the capture oligonucleotide sequences.
  • DESCRIPTION OF THE FIGURES
  • FIG. 1A-1E are representative flow cytometry histograms for positively validated anti-CD3-o20b(+)act and anti-CD28-o20b(+)act conjugates binding to T cells. 1A: Binding of a DNA (o20b(+)act) attached anti-CD28 antibody to T cells, 1B: Binding of corresponding unconjugated anti-CD28 antibody (positive control), 1C: Binding of a DNA (o20b(+)act) attached anti-CD3 antibody to T cells, 1D: Binding of corresponding unconjugated anti-CD3 antibody (positive control), and 1E, Cell incubated with labeled secondary antibody (negative control).
  • FIG. 2 is an illustration of the preparation of single stranded rolling circle amplification (ssRCA) to produce RCA products (RCAact) for T cell activation in two steps.
  • FIG. 3A is graphical representation of T cell activation measured as population of cells expressing CD25 receptor (a marker of T cell activation) by DBTA (DNA-Based T cell Activation) relative to the CD3/CD28 Dynabeads benchmark with respect to both early (days 1 and 4) % CD25 expression. Also shown are various controls including unconjugated antibodies alone and cells alone (without activator) not expected to cause significant activation.
  • FIG. 3B is a graphical representation of the same cultures as in FIG. 3A at days 4 and 7 showing cell expansion represented as number of folds of expansion relative to starting count (cell counts).
  • FIG. 4A is a graphical representation showing significant activation (relative to control) achieved with different DBTA systems, a DBTA system wherein only the anti-CD3 antibody is attached to the o20b(+)act (anti-CD3-o20b(+)act) and another DBTA system wherein both anti-CD3 and anti-CD28 are attached to o20b(+)act. Measured is the % CD25 expression.
  • FIG. 4B is a graphical representation of the same cultures as FIG. 4A after 4 and 7 days showing x-fold expansion (cell counts) as in FIG. 3B.
  • FIG. 5A is a graphical representation showing a trend towards higher activation when the DBTA components are mixed in situ compared to when they are pre-incubated/pre-associated and then mixed with cells.
  • FIG. 5B is the same cultures as FIG. 5A after 4 days showing x-fold expansion (cell counts).
  • FIG. 6A is a graphical representation showing T cell activation (24-hour % CD25) under a variety of conditions including different ratios of anti-CD3-DNA and anti-CD28-DNA with same RCA amount (10 RCA repeat units/per anti-CD3-DNA conjugate) and different amounts of DBTA reagent while keeping the ratio of anti-CD3-DNA conjugate and RCA repeat unit constant at 1:10.
  • FIG. 6B is a graphical representation of cell expression of the same cultures as FIG. 6A after days 4 and 7 showing x-fold expansion (cell counts).
  • FIG. 7A is a graphical representation of greater than two-fold lower early activation (24-hour % CD25 expression) for sample where cells were pre-mixed with anti-CD3 and anti-CD28 DNA conjugates for 30 minutes prior to adding RCA compared to all three DTBA components added at once (one after another).
  • FIG. 7B is a graphical representation of the same cultures as FIG. 7A showing diminished day 7 cell expansion (x-fold expansion, cell counts).
  • FIG. 8A is a graphical representation showing T cell activation (% CD25 expression) is achieved for both CpG and no CpG RCA at different ratios (1:1, 1:10 and 1:100) of the anti-CD3-DNA:RCAact.
  • FIG. 8B is a graphical representation of the same cultures as FIG. 8A showing x-fold expansion (cell counts) after 4 and 7 days.
  • FIG. 9A is a graphical representation showing T cell activation with a different DNA sequence (a portion of MRSA (Methicillin-resistant Staphylococcus aureus) sequence) attached to the anti-CD3 antibody and its complementary RCA product.
  • FIG. 9A is a graphical representation of the same cultures in FIG. 9A showing x-fold cell expansion after days 4 and 7.
  • FIG. 10A is a graphical representation showing that the larger the RCA product, the higher the activation (% CD25 expression).
  • FIG. 10B is a graphical representation of the same cultures in FIG. 10A showing higher x-fold expansion (cell counts) with larger product in early expansion phase (day 4), however, by day 7 all the three RCA products of different sizes yield comparable levels of cell expansion.
  • DETAILED DESCRIPTION
  • The singular forms “a” “an” and “the” include plural referents unless the context clearly dictates otherwise. Approximating language, as used herein throughout the specification and claims, may be applied to modify any quantitative representation that could permissibly vary without resulting in a change in the basic function to which it is related. Accordingly, a value modified by a term such as “about” is not to be limited to the precise value specified. Unless otherwise indicated, all numbers expressing quantities of ingredients, properties such as molecular weight, reaction conditions, so forth used in the specification and claims are to be understood as being modified in all instances by the term “about.” Accordingly, unless indicated to the contrary, the numerical parameters set forth in the following specification and attached claims are approximations that may vary depending upon the desired properties sought to be obtained by the present invention. At the very least each numerical parameter should at least be construed in light of the number of reported significant digits and by applying ordinary rounding techniques.
  • While certain embodiments are directed towards autologous cell therapies that involve collection, manipulation, and re-insertion of a patient's own cells, the applications of the disclosed techniques may include allogenic cells, modified human cells, or xenotransplantation of non-human cells. Cell based therapies that are contemplated as being used in conjunction with the disclosed techniques may include therapies for organ or tissue regeneration, cancer treatment, blood disorders, immunotherapies, heart disease, or any other cell-based therapies. A variety of cell types may be utilized within the context of the present invention including for example, but not limited to, cell types such as B cells, T-cells, or natural killer cells. The cells can be isolated from any tissue such as peripheral blood, bone marrow, or tumor tissue. Some embodiments are directed towards T cells enriched from peripheral blood by centrifugation using for example Ficoll-Paque™ or Percoll™ (GE Healthcare) gradient. Some other embodiments are directed towards a specific subpopulation of T-cells, such as CD28+, CD4+, CD8+, CD45RA+, and/or CD45RO+ T cells, further isolated by positive or negative selection techniques.
  • As used herein “second agent” refers to a polymer comprising a plurality of capture oligomers, wherein at least a segment of at least one of the plurality of capture oligomers is capable of associating with a first binder moiety. In some embodiments the second agent is a nucleic acid polymer such as a rolling circle amplification product and the “capture oligomer” is a capture oligonucleotide sequence. The nucleic acid polymer may comprise a plurality of the same or different capture oligonucleotide sequences. In some embodiments the number of capture oligonucleotide sequences in the nucleic acid polymer is greater than 3, preferably greater than 30, and more preferably greater than 300. In certain embodiments, the number of capture oligonucleotide sequences is greater than 2000. As such, the nucleic acid polymer may comprise a plurality of capture oligonucleotide sequences of the same or different nucleotide sequence, molecular weights, geometrical arrangements, and/or patterns of repetition. In some embodiments, the capture oligonucleotide sequence may have a length in range of from about 6 nucleotides to about 12 nucleotides, from about 12 nucleotides to about 25 nucleotides, from about 25 nucleotides to about 50 nucleotides, from about 50 nucleotides to about 100 nucleotides, from about 100 nucleotides to about 250 nucleotides, or from about 250 nucleotides to about 500 nucleotides. In some agents the second agent is double-stranded nucleic acid polymer while in other embodiments it is a single-stranded nucleic acid polymer. The nucleic acid polymer may include natural or unnatural nucleotides with modifications on the nucleic acid base, sugar or phosphate backbone. The second agent may further comprise spacer oligonucleotide sequences having different number and/or sequence of nucleotides than the capture oligonucleotide sequences, or non-nucleic acid spacer molecules. In some other embodiments the second agent is a cationic polymer comprising a plurality of capture cationic oligomers, containing contiguous stretches of cationic monomers such as histidine or lysine. The second agent may further comprise neutral or anionic spacer residues provided the overall charge remains cationic. In still other embodiments the second agent is an anionic polymer comprising a plurality of capture anionic oligomers, containing contiguous stretches of anionic monomers such as acrylate. The second agent may further comprise neutral or cationic spacer residues provided the overall charge is anionic.
  • As used herein, the term “first agent” refers to a cell activator attached to a “first binder moiety”. In some embodiments, the cell activator is a T-cell activator. The term “first binder moiety” refers to a molecule that can associate with one or more capture oligomers. In some embodiments the first binder moiety is a nucleic acid sequence and the “second agent” is a nucleic acid polymer comprising a plurality of capture oligonucleotide sequences, such that the nucleic acid sequence of the first binder moiety is complementary to at least a segment of at least one of the plurality of capture oligonucleotide sequences. The nucleic acid sequence of the first binder moiety and the capture oligonucleotide sequence are capable of associating via complementary base-pair hybridization. The first binder moiety and the capture oligonucleotide may hybridize to form a double or triple helix structure depending on whether the nucleic acid polymer is a double-stranded nucleic acid polymer or a single-stranded nucleic acid polymer. In some embodiments, all the nucleotide residues of at least one capture oligonucleotide sequence may hybridize to complementary nucleotides in the first binder moiety. For example, at least one capture oligonucleotide sequence may have 50 nucleotide residues and all the 50 nucleotide residues may hybridize to complementary nucleotides in the first binder moiety. In some embodiments, all the nucleotides of the capture oligonucleotide sequence may not have corresponding complementary nucleotides in the nucleic acid sequence of the first binder moiety. In some embodiments, there may be one or more base-pair mismatches between the capture oligonucleotide sequence and the nucleic acid sequence of the first binder moiety. In still other embodiments the capture oligonucleotide sequence and/or the nucleic acid sequence of the first binder moiety may include nucleic acid analogs, with modified bases such as azidothymidine, inosine, or uridine, or modified sugars, e.g. 3′-OMe, or modified backbone, e.g. phosphorothioate, alkylphosphonate, phosphoramidate backbones. The first or second agent may further comprise non-complementary spacer oligonucleotide sequences or non-nucleic acid spacer molecules. In some other embodiments the first binder moiety is an anionic moiety such as a nucleic acid sequence, alginate, polyglutamate, polyaspartate or hyaluronate and the second agent is a polymer comprising a plurality of cationic capture oligomers. The anionic first binder moiety is capable of associating with at least a segment of at least one of the plurality of cationic capture oligomers via electrostatic association. In still other embodiments, the first binder moiety is a cationic moiety and the second agent is a polymer comprising a plurality of anionic capture oligomers.
  • As used herein, the term “T-cell activator” refers any agent that can activate a T-cell for proliferation and/or transduction. Suitable examples of T-cell activator include small organic molecules (for example ionomycin, phorbol myristate or acetate), natural or modified peptides, proteins (for example antibodies or affibodies), non-natural peptide mimics, nucleic acids (for example polynucleotides, PNA, DNA, RNA or aptamers), polysaccharides (for example lectins or sugars), lipids, enzymes, enzyme substrates or inhibitors, ligands, receptors, antigens, or haptens. In some embodiments, the T-cell activator binds to a T-cell surface receptor, such as a T-cell activating receptor, which results in delivery of a primary activation signal in a T-cell. T-cell activating receptors include for example T-Cell Receptor (TCR) or CD3 receptor. A primary activation signal can be initiated through binding between a TCR and an antigen presented in conjunction with either MHC class I or class II molecules, in order to stimulate an antigen-specific T-cell activation. A primary activation signal can also be initiated through binding between a CD3 receptor and a ligand targeted to the CD3 receptor, in order to stimulate a polyclonal T-cell activation. In exemplary embodiments the T-cell activator is an anti-CD3 antibody or fragments thereof. Other examples of T-cell activator include concanavalin A, protein kinase C (PKC) activator such as a phorbol ester (for example phorbol myristate acetate) or calcium ionophore (for example ionomycin which raises cytoplasmic calcium concentrations) and others. In some embodiments a first agent that binds to a TCR or a CD3 receptor may be used in conjunction with other T-cell activators.
  • In some embodiments, the method of activating T cells comprises the addition of a plurality of a 1st population of first agents, wherein all the first agents in the 1st population of first agents comprises T-cell activator of the same type, such as an anti-CD3 antibody. In some embodiments, the method may further comprise the addition of a 2nd, 3rd, . . . nth population of first agents. The T-cell activators among different populations of first agents may be of the same type or different types such as anti-CD3 antibodies or fragments thereof or antigens bound to MHC molecules.
  • In some embodiments, the method of activating T cells further comprises the addition of a T-cell co-stimulator. Examples of T-cell co-stimulators include ligands targeted towards T-cell co-stimulatory receptors such as CD28, CD2, ICOS, OX40, or 4-IBB receptors. Suitable ligands may include one or more of natural or modified peptides, proteins (for example antibodies, affibodies), non-natural peptide mimics, nucleic acids (for example polynucleotides, PNA, DNA, RNA, or aptamers), polysaccharides (for example lectins, sugars), lipids, enzymes, enzyme substrates or inhibitors, ligands, receptors, antigens, or haptens. The addition of a T-cell co-stimulator may include the addition of one or more types of T-cell co-stimulators. In some embodiments the T-cell co-stimulator may be attached to a second binder moiety. The term “second binder moiety” refers to a molecule that can associate with one or more capture oligomers. In some embodiments, the second agent is a nucleic acid polymer and the second binder moiety is a nucleic acid sequence, which is same as or different from the nucleic acid sequence of the first binder moiety. In some other embodiments the second agent is a cationic polymer and the second binder moiety is an anionic moiety. In still other embodiments the second agent is an anionic polymer and the second binder moiety is a cationic moiety.
  • The term “attached” refers to any means of attachment known in the art. A T-cell activator can be attached to a first binder moiety by any means provided the attachment does not interfere with the activator's ability to activate T cells. Similarly, a T-cell co-stimulator can be attached to a second binder moiety by any means provided the attachment does not interfere with the co-stimulator's ability to provide a co-stimulatory signal to the T cell. The attachment may be covalent or noncovalent, electrostatic, or hydrophobic. In a preferred embodiment the attachment is covalent.
  • In a preferred embodiment the T-cell activator is an anti-CD3 antibody attached to a nucleic acid sequence, the T-cell co-stimulator is an anti-CD28 antibody attached to a nucleic acid sequence, and the second agent is a rolling circle amplification product comprising a plurality of complementary capture oligonucleotide sequences.
  • In preferred embodiments the first agent and the second agent are soluble in cell culture media. As to be understood, “cell culture media” refers to any standard T cell culture media, which are known in the art. Illustrative media include but are not limited to RPMI 1640, AIM-V, DMEM, MEM, α-MEM, F-12, X-Vivo 15. The T cell culture media may be serum-free or supplemented with serum, such as human serum or serum supplement. In some embodiments, the T cell culture media is further supplemented with additional growth factors and cytokines such as interleukin-2 (Il-2), interleukin-7 (Il-7) or interleukin-15 (IL-15).
  • As used herein, the term “in situ” generally refers to an event occurring in the original location, for example in the location where T cell activation occurs. In some embodiments “in situ” refers to an event happening in the T cell culture media under cell culturing condition.
  • The invention includes embodiments that relate generally to methods of activating T cells for cellular therapy. In some embodiments, the method includes the addition of a plurality of first agents, each first agent comprising a T-cell activator attached to a first binder moiety, wherein the T-cell activators are capable of binding to a set of T-cell surface receptors, such as the T-cell activating receptors, inducing a cellular event within the T cells that leads to activation and proliferation of the cells. In some embodiments a clustering of the T-cell surface receptors is induced by a second agent, which associates with the first binder moiety bringing the T-cell activators in proximity to each other. In some embodiments, the method further includes the addition of a plurality of T-cell co-stimulators capable of binding to a different set of T-cell surface receptors, such as the T-cell co-stimulatory receptors. The T-cell co-stimulator may be attached to a second binder moiety. The second agent associates with both the first binder moiety and the second binder moiety thereby inducing a clustering of the T-cell surface receptors. In some embodiments the second agent is a nucleic acid polymer comprising a plurality of capture oligonucleotide sequences and the association is caused by base-pair hybridization between a nucleic acid sequence attached to the T-cell activator and a complementary capture oligonucleotide sequence. In some embodiments the method further comprises the addition of a T-cell co-stimulator attached to a second binder moiety, wherein the second binder moiety is also a nucleic acid sequence, and the nucleic acid sequence is capable of association with a complementary capture oligonucleotide sequence by base-pair hybridization. In some embodiments the association of the capture oligomer with the first binder moiety and the second binder moiety is caused by electrostatic interaction between oppositely charged species for example anionic moieties attached to the T-cell activator and T-cell co-stimulator with cationic capture oligomers or vice versa.
  • In some embodiments, the first agent, the second agent, and optionally a T-cell co-stimulator, which may or may not be attached to a second binder moiety, are added simultaneously or sequentially in any order, to a population of T cells provided in a cell culture media, to allow an in-situ association. In still other embodiments, the first agent, the second agent, and optionally a T-cell co-stimulator attached to a second binder moiety, are allowed to pre-incubate and associate to form a pre-associated complex. The pre-associated complex is then added to the population of T cells. In preferred embodiments, the first agent, the second agent, and optionally the T-cell co-stimulator attached to a second binder moiety, are allowed to associate in-situ.
  • The T cells are incubated with the first agent, the second agent, and optionally a T-cell co-stimulator for a period of time, whereby at least a portion of the population of T cells is activated. In some embodiments the T-cell co-stimulator is attached to a second binder moiety. In some embodiments the time sufficient to activate a portion of the population of T cells may range from about 1 minute to about 14 days. In certain embodiments, the time may be from about 24 h to about 8 days. In some embodiments, at least 15% of T cells are activated. In preferred embodiments at least 25% of the T cells are activated, and in more preferred embodiment the majority of the T cells, greater than 50% are activated. The incubation may be done in a bioreactor having a controlled environment of temperature, humidity, CO2 concentration, for example at 37° C. and 5% CO2. In some embodiments the incubation may be done in a static bioreactor where there is no movement of the bioreactor, while in some other embodiments the incubation may be done in a bioreactor placed on a rocking platform.
  • In some embodiments the invention provides methods to activate and selectively expand a specific subpopulation of T cells from a mixed population of T cells. This can be achieved for example by varying the nature or relative proportion of the T-cell activators and T-cell co-stimulators. Further, the invention provides methods to control T-cell surface receptor clustering and hence activation by adjusting for example the number of or distance among the T-cell activators and T-cell co-stimulators, which are associated with the second agent. In an exemplary embodiment where the second agent is a nucleic acid polymer, this can be achieved by precisely controlling the number and/or length of the capture oligonucleotides, or the length of the spacers.
  • The expression of certain antibodies such as CD25 may be used to measure the activation of the T cells. In certain embodiments the expression of CD25 receptors on the T cells may be assayed by labeled anti-CD25 antibodies to enumerate the labeled cells. In certain other embodiments, activation may be measured by other markers signifying T-cell surface receptor clustering, which induces activation and expansion. The other markers used to measure T cell activation and expansion include CD4, CD8, CD27, CD28, CD3, CD57, CD25, and CD62L.
  • In preferred embodiments, the first agent and the second agents are bio-degradable. As used herein, the term bio-degradable refers to materials that degrade in biological fluid. In some embodiments, the degradation may occur using chemical or enzymatic means. In some embodiments, where the second agent is a nucleic acid polymer and/or the first binder moiety is a nucleic acid sequence, the method of activation of T cells further comprises the addition of a degrading enzyme such as a nuclease. In some embodiments the degrading enzyme, such as the nuclease, is added after at least a portion of the population of T cells is activated. In some embodiments, the degrading enzyme is added after the T cells have expanded at least 10-100 fold. In still other embodiments, the degrading enzyme is added at the end of the culture period, before the T cells are harvested from the cell culture media, prior to washing, concentration and final formulation. It should be noted that the degrading enzyme can be added any time after at least a portion of the population of T cells is activated and before the T cells are administered into a patient. In certain embodiments, after degradation, the degraded by-products of the first and second agents can be removed during washing and concentration of the T cells, and additional purification steps are not necessary. In some other embodiments, the second agent and the first agent are biocompatible and may be rapidly degraded in the blood stream. In such cases, the addition of a degrading enzyme may not be required before the T cells are administered into a patient. The use of such soluble, biodegradable systems are advantageous over polystyrene beads or other comparable bead based approaches since the use of such systems may avoid the need for additional purification steps, such as magnetic separation, which often leads to significant cell loss. In embodiments that include the addition of a T-cell co-stimulator attached to a second binder moiety, the second binder moiety is also biodegradable.
  • In some embodiments, the method of T cell activation further comprises the addition of a vector comprising a foreign gene. In some embodiments the foreign gene encodes a chimeric antigen receptor or a T-cell receptor. In some embodiments, the vector is a viral vector such as a γ-retroviral vector or a lentiviral vector. In some other embodiments the vector is a plasmid vector. In some embodiments the vector is added simultaneously with the first agent, while in some other embodiments, the vector is added after at least a portion of the population of T cells is activated.
  • Some embodiments are directed towards a kit comprising a T-cell activator attached to a nucleic acid sequence, a nucleic acid polymer comprising a plurality of capture oligonucleotide sequences, and optionally a T-cell co-stimulator. In some embodiments the T-cell co-stimulator is attached to a nucleic acid sequence, which is same as or different from the nucleic acid sequence attached to the T-cell activator. In specific examples the kit is a DNA-Based T cell Activation (DBTA) construct comprising a DNA polymer such as a rolling circle amplification product and one or more of the following components, i) a T-cell activator attached to a DNA sequence (first binder moiety), and ii) a T-cell co-stimulator attached to a DNA sequence (second binder moiety), which is same as or different from the DNA sequence attached to the T-cell activator.
  • EXAMPLES
  • The following examples are intended only to illustrate methods and embodiments in accordance with the invention, and as such should not be construed as imposing limitations upon the claims.
  • Example 1: Synthetic Preparation of DNA Attached Antibody (Ab-DNA) Conjugates for T Cell Activation
  • DNA-Based T cell Activation (DBTA) polymer constructs are representative nucleic acid polymer systems that initiate T cell clustering, activation, and subsequent expansion. DBTA comprises a DNA polymer produced via rolling circle amplification (such as RCAact) and consisting of repeating 43-base oligonucleotide segments and one or more of the following components: (1) anti-human CD3 monoclonal antibodies conjugated to 20-base DNA sequence derived from human beta-actin (o20b(+)act), (2) anti-human CD28 monoclonal antibodies also conjugated with DNA sequence o20b(+)act. The 43-base oligonucleotide segments bear full complementarity to the DNA sequences conjugated to the antibodies (1) and (2). The sequences of RCAact are derived from the complement to the RCA template sequences given below in Table 1:
  • TABLE 1
    Modification key: Phos = 5′ phosphate, Mal = maleimide functional group, C6 =
    hexylamino modification
    Sequence (5′→
    length 3′) including SEQ ID
    Name sense (b) modifications NO use/application
    RCA primer - 20 TGA CTA TTA 1 primer for RCA reactions, sequence derived
    AGA CTT CCT GT from human beta-actin
    RCA + 43 /Phos/TTA ATA 2 template for RCA reactions, sequence
    template GTC ATT CCA derived from human beta-actin and
    (CpG) AAT ATG AGA contains CpG island
    TGC GTT GTT
    ACA GGA AGT C
    RCA + 43 /Phos/TTA ATA 3 template for RCA reactions, sequence
    template GTC ATT CCA derived from human beta-actin and does
    (no CpG) ACA TAT GAG not contain CpG island
    ATG GTT GTT
    ACA GGA AGT C
    o20b(+)act + 20 /MalC6/ACA 4 conjugation to Ab via 5′-maleimide and
    GGA AGT CTT binding to human B-actin-derived RCA
    AAT AGT CA product
  • Synthesis of Anti-CD3-DNA and Anti-CD28-DNA Conjugates
  • The covalent attachment of DNA sequences to antibodies (Abs) proceeds via a maleimide-thiol coupling strategy as described below. The following specific description of anti-CD3-o20b(+)act and anti-CD28-o20b(+)act conjugate synthesis, purification, and characterization can be adapted as a general conjugation strategy for different antibody (Ab) clones as well as nucleic acid sequences of different length and composition.
  • Preparation of Maleimide-Activated DNA Sequences
  • The starting nucleic acid sequence (prot-mal-o20b(+)act) is provided by TriLink Biotechnologies and comprises a 20-base DNA sequence capped with an N-terminal C6 spacer followed by a protected tricyclic maleimide moiety. Upon receipt of this commercial material, a reactive mal-o20b(+)act derivative is generated via an inverse electron-demand Diels-Alder deprotection step, whereby the starting nucleic acid sequence is suspended in anhydrous toluene (˜1 mg/mL) and heated at 90° C. for 4 h. Precipitation of the nucleic acid sequences via benchtop centrifugation and removal of solvent is followed by several washes (3×1 mL) with cold ethanol. Upon further reduction of residual organic solvents under reduced pressure, the washed solid product is dissolved in 100 mM HEPES buffer, pH 7.3 and the solution concentration is determined via UV-Vis spectroscopy (NanoDrop). The resulting stock solution (0.5-1 mM in DNA) or a portion thereof may be used directly for antibody conjugation with thiol-modified antibody. The remaining mal-o20b(+)act stock may be stored for several months at −20° C. without significant loss of reactivity.
  • Preparation of Thiol-Modified Ab Intermediates.
  • A 10 mM stock solution of Traut's reagent (2-aminothiolane hydrochloride) is first prepared in 100 mM HEPES buffer, pH 7.3. A 1 mg/mL solution of anti-CD3 Ab (OKT3 clone, eBioscience) or anti-CD28 Ab (9.3 clone, GeneTex) in PBS is then mixed with both 20× pH 8.5 borate buffer (ThermoFisher) and 10 mM Traut's reagent stock solution in a 8:1:1 ratio by volume. The resulting solution is thoroughly mixed and allowed to incubate at room temperature for 0.75 h. The unused portion of the Traut's mixture may be stored for several months at −20° C. without significant loss of reactivity. Following antibody activation, the reaction mixture is purified using a conventional desalting column (e.g. NAP-5 or PD-10, GE Healthcare Life Sciences) that has been equilibrated with 100 mM HEPES, pH 7.3 buffer. The collected fractions are then immediately analyzed by UV-Vis spectroscopy (NanoDrop). The resulting protein recovery at this stage is typically >60% for both anti-CD3 and anti-CD28 antibodies using known molar extinction coefficients for antibody at 280 nm.
  • Conjugation of Maleimide-Activated DNA Sequences with Thiol-Modified Ab Intermediates to Generate Ab-DNA Conjugates
  • For the final Ab-DNA attachment step, the volume of mal-o20b(+)act corresponding to a target molar input ratio of 10-40:1 o20b(+)act:Ab is added to an aliquot of freshly prepared, purified, thiol-activated Ab. After thorough mixing, the resulting solution is allowed to incubate at room temperature overnight (16-24 h). Final conjugate purification is achieved via selective precipitation of Ab using a saturated ammonium chloride solution. First, a volume of saturated ammonium chloride equal to the total reaction volume is added, thoroughly mixed, and placed on ice. After 15 min, the sample is centrifuged at 15,000×g rcf for 10 min at 10° C. Removal of the supernatant is followed by addition of an appropriate minimum volume of 0.1M sodium phosphate, 0.15M NaCl, pH 7 buffer to re-dissolve the final pellet. The final antibody-DNA conjugate recovery and labeling efficiency (attached Ab-DNA) is determined using the Pierce BCA Protein Assay Kit (Thermo Scientific) in combination with NanoDrop A260 measurement for determination of DNA content (ε=210,100 M−1 cm−1). Under these condition an average of 1-3 o20b(+)act oligonucleotide molecules are conjugated to each molecule of anti-CD3 or anti-Cd28 antibody. Attachment is generally achieved with a final conjugate recovery of >60%. Further confirmation of conjugate purity is determined using analytical size exclusion chromatography (SEC) against a standard protein size calibration curve. Typical analytical SEC conditions are as follows: 10 μL sample injection volume, 0.5 mL/min flow rate, 30 min run using 100 mM sodium phosphate, 100 mM sodium sulfate, 0.05% sodium azide, pH 6.7 buffer on a TSK Gel 3000SW×L column (TOSOH Bioscience). Typical analytical SEC elution times are as follows: unlabeled Ab=16.8-17.0 min, starting o20b(+)act=20.0 min, Ab-DNA (Ab-o20b(+)act) conjugate mixture=10-15 min. Purified final Ab-DNA conjugates (after precipitation and resuspension) show >95% removal of unbound DNA intermediate or starting material upon SEC analysis.
  • Example 2: Validation of T Cell Binding for Anti-CD3-DNA and Anti-CD28-DNA Conjugates
  • To ensure that the Ab-DNA conjugates prepared in Example 1 retain their specific cell binding capabilities to T cells, validation studies are performed for each conjugate batch using flow cytometry (Cytoflex S, Beckman Coulter). NBP Pan T Cells (ALL Cells) are thawed at 37° C. in 10 mL warm complete media (see Example 4) and then centrifuged at 300×g rcf for 10 min. Cells are then resuspended in 10 mL fresh complete media and analyzed on a Nucleocounter® NC-200 system to determine cell counts and viability. After adjusting the concentration to 1×106 cells/mL and washing with PBS, the T cells are then blocked in 10% Normal Goat Serum (NGS) in PBS at 4° C. for 15 min. After removal of the blocking solution, cells are then incubated with primary antibody in 1% NGS/PBS solutions for 15 min at 4° C. In parallel, both anti-CD3-DNA and/or anti-CD28-DNA conjugate (anti-CD3-o20b(+)act and/or anti-CD28-o20b(+)act) test samples (Example 1) as well as unlabeled or unconjugated anti-CD3 and/or anti-CD28 Ab positive controls (Ab not attached to DNA) are used. After primary Ab incubation, cells are washed in PBS and incubated with a 1% NGS/PBS solution of fluorophore-labeled secondary antibody (Jackson Immuno) specific for the mouse isotype of both anti-CD3 and anti-CD28 Abs. Typical dilutions for secondary antibody labeling are 1:200 of a 1 mg/mL stock solution. After 15 min incubation at 4° C., cells are washed as before, resuspended in PBS and analyzed by flow cytometry to determine the percentage of T cells bound with Ab-DNA conjugate relative to the percentage of cells bound with unlabeled Ab (positive control).
  • Representative flow cytometry histograms for positively validated anti-CD3-o20b(+)act and anti-CD28-o20b(+)act conjugates binding to T cells are shown in FIG. 1A through 1E; Anti-CD28-o20b(+)act, anti-CD28 unlabeled, Anti-CD3o20b(+)act, anti-CD3 unlabeled, and FITC-labeled respectively. For these particular Ab-DNA conjugate batches, high percentage of anti-CD3+ and anti-CD28+ cell binding is observed (>85% for both). These results indicate that despite DNA attachment, a significant portion of the anti-CD3 and anti-CD28 Ab samples retain their T cell binding capabilities.
  • Example 3: Production of a Nucleic Acid Polymer for T Cell Activation Using Rolling Circle Amplification
  • Preparation of single stranded rolling circle amplification (ssRCA) to produce RCA products (RCAact) for T cell activation comprises two steps: 1) ligation of a linear DNA strand to generate a circular template, and 2) amplification of the ligated circle to synthesize long single stranded concatemers. The ligation was accomplished using a high concentration of T4 DNA Ligase and its corresponding reaction buffer (New England Biolabs®) Two deoxyoligonucleotides (oligonucleotide) were present in the ligation. One oligonucleotide (RCAact-NoCG) consisted of 43 bases of the DNA sequence of the human β-actin gene and contained a 5′ phosphate group. This 43 base oligonucleotide would become the circular template following ligation. The second oligonucleotide was 20 bases in length (RCAact Primer) and was complimentary to both ends of the 43 base oligonucleotide. This second oligonucleotide was used to both form the circle prior to ligation and then amplify the circle in the subsequent ssRCA reaction (see FIG. 2). 450 pmol of the 43 base oligonucleotide were mixed in a 120 μL volume with 300 pmol of the 20 base oligonucleotide in an annealing buffer consisting of 10 mM Tris, pH 8, and 50 mM sodium chloride. The mixture was heated at 95° C. for two minutes and then cooled to +4° C. by dropping the temperature 0.1° C. every second. After cooling the mixture was warmed to room temperature and 96 μL of this annealing reaction was mixed with 48 μL of 10× T4 DNA Ligase Buffer containing 10 mM ATP and 24 μl of T4 DNA Ligase (400 units/μ1) in a final volume of 480 μL. The ligation reaction was allowed to incubate at 23° C. for 20 hours and then 65° C. for 20 minutes to heat-kill the ligase.
  • The ssRCA reaction was prepared by mixing 69.3 μL of the completed ligation reaction with 550 μL of 2× Phi29 Reaction Buffer (100 mM HEPES Buffer, pH 8.0, 150 mM potassium chloride, 2 mM TCEP, 40 mM magnesium chloride, 0.02% (v/v) Tween 20, 5% (v/v) polyethylene glycol and 1.6 mM each dATP, dCTP, dGTP and dTTP) in a final volume of 1.078 mL. After mixing, amplification was started by the addition of 22 μL of 1 mg/ml Phi29 DNA polymerase. Amplification reactions were incubated at 30° C. for 18 hours and then 65° C. for 15 minutes to heat-kill the polymerase. Completed ssRCA reactions were split equally into three separate tubes and precipitated by the addition of 0.1 volume 3M sodium acetate and 2.5 volumes of 95% (v/v) ethanol. Precipitations were allowed to stand at room temperature for 30 minutes and then centrifuged at high speed (>20K×g) for 30 minutes. The supernatants were removed by aspiration, each DNA pellet rinsed with 500 μl of 70% (v/v) ethanol and then recentrifuged at high speed (>20K×g) for 5 minutes. The supernatants were again removed by aspiration and the DNA pellets resuspended in TET Buffer (10 mM Tris, pH 8.0, 0.1 mM EDTA and 0.01% (v/v) Tween 20).
  • The apparent size of RCAact products is determined by pulse field gel electrophoresis relative to yeast (S. cerevisiae) or lambda DNA molecular weight ladders. In addition to pulse-field analysis, gel-shift assays using non-denaturing agarose gels are used to confirm hybridization of anti-CD3-o20b(+)act and anti-CD28-o20b(+)act conjugates (Example 1) with RCAact.
  • Example 4: Comparison of T Cell Activation and Expansion Using Dynabeads® Human T-Expander CD3/CD28 and a DBTA System of Nucleic Acid Polymers with Ab-DNA Conjugates (Anti-CD3-DNA and Anti-CD28-DNA)
  • Frozen aliquots of human Pan T Cells from AllCells (Catalog# PB009-IF) are used for all activation and expansion studies. Pan T Cells are thawed and processed as described in Example 2 and added to following complete X-Vivo media to give an initial concentration of 1×106 cells/mL as shown in Table 2.
  • TABLE 2
    X-Viro media aliquots
    Final
    concen-
    tration Volume
    Component Vendor Cat# in media (ml)
    Human serum off Valley HS1017 5% 50
    the clot Biomedical
    Glutamax 1-CTS Gibco A12860-01 1% 10
    Pen-Strep ThermoFisher 15140-122 1% 10
    N-acetyl cysteine Sigma A9165 0.8%   8
    IL-2 Thermo Fisher 200-02 200 IU 0.152
    X-Vivo media Lonza BE04-743Q 92%  1000
  • Typical activation and expansion experiments are performed in a 6-well format using a 2 mL seeding volume per well with a minimum of one additional replicate per condition tested. Dynabeads® Human T-Expander CD3/CD28 (Catalog#111.41D, ThermoFisher) are prepared according to the manufacturer's instructions. Briefly, a bead aliquot is washed with 0.3 mL complete media three times, with supernatant wash removal occurring after sample application to DynaMag-2 permanent magnet (ThermoFisher). After resuspension in complete media, a 60 bead slurry aliquot is added per well such that an approximate 3:1 bead-to-cell ratio is used for initial activation conditions (starting with 2×106 total cells per well).
  • Unless otherwise specified, aliquots of DBTA components (Ab-DNA and RCAact) are added separately and consecutively to cells directly from their respective stock solutions (stored at 4° C.). Additions of each DBTA component may be made in any sequential order. Standard initial concentrations of DBTA components are as follows: 1 μg/mL (6.7 nM) each for anti-CD3-o20b(+)act (anti-CD3-DNA) and anti-CD28-o20b(+)act (anti-CD28-DNA) with 10-fold molar excess of RCAact (˜67 nM). RCAact concentration is based on the molar concentration of the repeating 43-base segment and independent of total RCA product length or polydispersity. Following the addition of all of the appropriate activating components, the well contents are mixed with a 1 mL pipette. The plates are then incubated at 37° C. and 5% CO2 atmosphere under static conditions for 1-7 days or longer with periodic aliquots taken and, dilutions done with fresh media as needed for continuing expansion and analysis. Confirmation of cell activation is achieved by % CD25 expression using flow cytometry after 24 h incubation (day 1) and followed by additional measurements, if needed, at days 4 and 7. Cell counts, viability, and size (blasting) are additionally measured at days 4 and 7 using the Nucleocounter. Based on day 4 cell counts, a 1:4 or 1:8 dilutions with fresh complete media is performed to reduce cell density to 250,000 cells/mL per well. This enables cells to expand in the exponential phase, without overgrowth, until day 7 analysis. For the negative control (“cells-only”) sample group in which no activating agents are added, cell density is maintained at a minimum of 500,000 cells/mL. Further phenotypic analysis via flow cytometry may be conducted at day 7 for select samples of interest. The panel of cell surface markers under investigation includes CD4, CD8, CD27, CD28, CD3, CD57, CD25, and CD62L. FIGS. 3A and 3B shows clear, comparable performance of DBTA relative to the CD3/CD28 Dynabeads benchmark with respect to both early (days 1 and 4) % CD25 expression as well as x-fold cell expansion after days 4 and 7. Control samples with unmodified antibodies, RCA alone, unmodified antibodies with unconjugated nucleic acid sequence (free oligo) and cells only show very low level of activation and expansion, as expected. The robustness of the DBTA system is also confirmed as the standard deviations of activation and expansion match those observed with Dynabeads. FIGS. 3A and 3B includes data from five separate experiments, seven individual human T cell donors, and 4 different batches of antibody conjugates. FIGS. 4A and 4B show that significant activation and expansion (relative to control) can also be achieved with a DBTA system wherein only the anti-CD3 antibody is attached to the o20b(+)act (anti-CD3-o20b(+)act). FIG. 4A shows the CD25 expression of the cultures after 24 hrs while FIG. 4B shows the CD25 expression of the same cultures after 4 and 7 days of expansion. However, the best performance is achieved when both anti-CD3 and anti-CD28 antibodies are attached to DNA to form the Ab-DNA conjugates. The data for FIG. 4 represents averages taken from over 5 separate experiments encompassing over six individual human T cell donors.
  • Example 5: Comparison of Large-Scale T Cell Activation and Expansion Using Dynabeads® (ThermoFisher Scientific, Waltham, Mass.) Human T-Expander CD3/CD28 and a DBTA System of Nucleic Acid Polymers (RCAact) with Ab-DNA Conjugates (Anti-CD3-o20b(+)Act and Anti-CD28-o20b(+)Act)
  • In addition to 6-well plate studies, a large scale comparison of T cell activation and expansion efficiency using 72C VueLife® bags (Cell Genix GmbH, Breisgau, Germany) was also undertaken. The same ratios and components described in Example 4 were scaled linearly to accommodate starting conditions of 32×106 cells in 32 mL of complete X-Vivo media (1×106 cells/ml). The cultures were maintained in the 72C VueLife bags within a standard cell culture incubator. On Day 4, cells were collected, washed with fresh media, counted, diluted in appropriate media volume to give 0.5×106 cells/mL and re-seeded within the VueLife 72C. On Day 6, cells were diluted and seeded in 250 mL in the Wave bag, on a rocking WAVE platform for further expansion. By Day 8, cells were counted using Nucleocounter® (ChemoMetec, Allerod, Denmark) and examined by flow cytometry for various CD surface marker expression. The tables below depict comparable levels of CD25 expression, x-fold expansion, cell size, and viability for both Dynabead samples and those activated with DBTA. In addition, the day 8 flow analysis shows nearly identical levels of phenotypic expression for the full panel of CD markers for both Dynabeads and DBTA. The preparation and results are illustrated in Tables 3-5 below.
  • TABLE 3
    Cell Counts at 4 and 8 days.
    Cell counts Fold X
    % CD25 Day 4 Day 4 (total Day 8 Day 8/
    Sample (24 h) (cells/ml) viable cells) (cells/ml) Day 4 Day 6
    DBTA >90% 8.45E+05 4.23E+07 1.57E+06 1.32 6.04
    Dynabeads >90% 1.25E+06 6.25E+07 1.38E+06 1.95 5.31
  • TABLE 4
    Cell Viability as a percentage of population.
    Day 8
    total Viable Cell size (μm) % Viability
    Sample Cells Day 4 Day 8 Day 4 Day 8
    DBTA 3.93E+08 12.2 10.9 98.1 97.5
    Dynabeads 3.45E+08 12.1 11.0 97.6 97.1
  • TABLE 5
    Cell Flow Cytometry results at 8 days.
    Flow Cytometry Results at day 8
    (% cells positive):
    CD Marker DBTA Dynabeads
    CD8 46% 45%
    CD4 51% 53%
    CD3 96% 98%
    CD25
    80% 84%
    CD57
    10%  8%
    CD62L 95% 96%
    CCR7 62% 47%
    CD28 96% 95%
    CD27 98% 96%
    CD27+CD28+ 90% 82%
  • Example 6: The Effect of Pre-Incubating DBTA Components Together Prior to Cell Culture Addition Versus Simultaneous Addition of all Components at the Start of Activation
  • A series of T cell activation experiments were conducted to assess the effect of pre-incubating all of the DBTA system components (Anti-CD3-DNA and anti-CD28-DNA conjugates along with their complementary RCAact polymeric product) and then adding the pre-associated complex to T cells. The results were compared to the standard protocol of simultaneous addition of each separate component to T cells for in-situ association. For these pre-incubated samples, the same quantities and ratios of DBTA components used in Example 4 were added together in a 1.5 mL tube, mixed thoroughly, and incubated at room temperature for 30 minutes. The pre-incubated DBTA mixture was then added to fresh T cell cultures in the 6 well plates. In parallel, and as conducted in Example 4, standard DBTA samples not subjected to pre-incubation were added to T cell cultures along with standard Dynabead and cells only samples (positive and negative controls, respectively).
  • FIGS. 5A and 5B shows that comparable levels of both early activation (24-hour % CD25 expression) and day 4 cell expansion is achieved whether or not the all the DBTA components are pre-incubated/pre-associated (anti-CD3-DNA/anti-CD28-DNA DBTA pre-associated) together or added simultaneously at the start of cell activation (anti-CD3-DNA/anti-CD28-DNA DBTA in-situ) FIG. 5A is after 24 hr incubation and 5B is after 4 days. Both sets of DBTA samples likewise compare favorably to benchmark Dynabead numbers for activation and early expansion. This data is derived from two separate experiments featuring four different human T cell donors and four different batches of Ab-DNA conjugates.
  • Example 7: The Effect of Different Input DBTA Antibody Conjugate and RCA Polymeric Product Quantities on T Cell Activation Efficiency
  • A series of T cell activation experiments were conducted to assess the effect of different ratios of Ab-DNA conjugate and RCAact on T cell activation efficiency. In all cases, the same anti-CD3-DNA: RCAact molar ratio of 1:10 was maintained, while 1:1 versus 1:2 molar ratios of anti-CD3-DNA: anti-CD28-DNA conjugate were examined. All other protocol conditions outlined in Example 4 were maintained.
  • FIG. 6A shows that sufficient T cell activation (24-hour % CD25 expression) and FIG. 6B cell expansion (days 4 and 7) are achieved at a variety of input quantities of antibody and RCAact product. Not surprisingly, the lowest input quantities of both antibody conjugate (0.25 μg/mL input or ˜170 pM) and the corresponding RCAact product (˜17 nM) showed the lowest levels of 24-hour % CD25 expression (60%), although this level of activation still far exceeds the cells-only negative control (20%) and eventually leads to day 7-fold expansion that matches the benchmark Dynabead samples. It is noted that half the standard input quantity of Ab-DNA conjugate (0.5 μg/mL) and RCAact (33.5 nM) nonetheless achieves the same level of early activation (90%) as the Dynabead benchmark. The data for this example is derived from three separate experiments featuring five different human T cell donors and four different batches of Ab-DNA conjugates.
  • Example 8: The Effect of Pre-Incubating DBTA Antibody Conjugates with Cells Prior to the Addition of RCA Polymeric Product for T Cell Activation
  • An experiment was conducted to assess the effect of adding the anti-CD3-DNA and anti-CD28-DNA antibody conjugates to T cell culture to generate a Ab-DNA/cell pre-incubated sample prior to a separate, second addition of RCA polymeric product 30 min later. For this experiment, all other protocol details and quantities outlined in Example 4 were followed.
  • FIG. 7A highlights a greater than two-fold lower early activation (24-hour % CD25 expression) and FIG. 7B shows diminished day 7 cell expansion for the Ab-DNA/cell pre-incubated sample (anti-CD3-DNA/anti-CD28-DNA DBTA (Ab-DNA/cells pre-incubated)) relative to both standard DBTA samples (featuring simultaneous addition of all components) (anti-CD3-DNA/anti-CD28-DNA DBTA in situ) and Dynabead controls. FIG. 7B also shows results of expansion after 4 days.
  • Example 9: The Effect of Different Input Ab-DNA Conjugate-to-RCA Product Ratios on T Cell Activation Efficiency
  • A series of T cell activation experiments were conducted to assess the effect of different Ab-DNA conjugate-to-RCAact input ratios on T cell activation efficiency. In all cases, only anti-CD3-DNA (anti-CD3-o20b(+)act) conjugates were used. Each Ab conjugate sample, furthermore, featured the same standard concentration per well for activation (1 μg/mL input or ˜6.7 μM), while a three log range of input RCAact product was investigated (6.7 nM, 67 nM, and 670 nM) to give starting Ab-DNA:RCA ratios of 1:1, 1:10, and 1:100, respectively. In addition, both CpG and non-CpG RCA template variants (see Example 1) were investigated. For these experiments, all other protocol details and quantities outlined in Example 4 were maintained.
  • FIGS. 8A and 8B shows that optimal T cell activation (% CD25 expression) is achieved for both CpG and no CpG RCA at the anti-CD3-DNA:RCAact ratio of 1:10. Diminished activation efficiency is observed at both higher and lower ratios of Ab-DNA-to-RCA. All samples, however, demonstrate day 7 T cell expansion of about 10-fold or greater, matching or exceeding the Dynabead control sample. FIG. 8A is after 1 and 4 day culture showing % CD25, FIG. 8B is after 4 and 7 days showing x-fold expansion.
  • Example 10: Demonstration of Human T Cell Activation and Expansion Using an Alternative DBTA System with a Different Sequence and Oligo Length
  • All previous examples (1-9) and associated FIGS. (1-8) for this disclosure successfully utilize a DBTA system featuring DNA sequences derived from human B-actin (Example 1). Successful human T cell activation and expansion may also be achieved with any number of alternative systems featuring different DNA sequences, base content, and oligo lengths. One alternative example features oligo and RCA products originally derived from the methicillin-resistant Staphylococcus aureus (MRSA) genome. The specific sequence information for each of these DBTA components is shown in Table 6 below:
  • TABLE 6
    Sequence Information.
    Sequence (5′→
    length 3′) including SEQ ID
    Name sense (b) modifications NO use/application
    RCA primer - 20 ATC AAT GAT 5 primer for RCA reactions, sequence derived
    (MRSA) GCA TAA CAT CT from MRSA genome
    RCA template + 43 /Phos/CAT CAT 6 template for RCA reactions, sequence derived
    (MRSA) TGA TTT AGA from MRSA genome
    CAC TGA AAA
    AGT TCG AGG
    AGA TGT TAT G
    o25b(+)mrsa + 25 /MalC6/CAT CAT 7 conjugation to Ab via 5'-maleimide and binding
    TGA TTT AGA to MRSA-derived RCA product
    CAC TGA AAA A
  • FIGS. 9 A and 9B shows clear, comparable performance of DBTA component (anti-CD3-DNA conjugate only with soluble anti-CD28 Ab) relative to the CD3/CD28 Dynabeads benchmark with respect to both early (days 1 and 4) % CD25 expression (FIG. 9A) as well as x-fold cell expansion after days 4 and 7 (FIG. 9B). These results are comparable to the β-actin derived DBTA system shown in the above examples. Also as with the β-actin DBTA, the specificity of the system is confirmed: control samples with unmodified antibodies, RCA product alone, and cells only show very low level of activation and expansion. It is noted that the exact same Ab conjugation, RCA production, and cell culturing protocols and molar quantities described above for β-actin DBTA were also utilized for the MRSA system.
  • Example 11: Demonstration of Human T Cell Activation and Expansion Using a DBTA System Featuring Cleaved RCA Polymeric Products
  • A comparison of human T cell activation and expansion efficiency using different sized RCA products was conducted using the MRSA-based DBTA system (See Example 10 and Example 3 for general conditions used to generate full length RCA product). For these experiments, full length RCA products were subjected to sonication conditions yielding different relative size distributions. To produce sonicated RCA products, a Covaris M220 Focused Ultrasonicator™ (Covaris, Woburn, Mass.) was utilized according to the manufacturer's recommended input conditions to yield 1500 bp and 150 bp sized fragments starting from large genomic DNA fragments. Target input parameters include peak incident power, duty factor, cycles per burst, treatment time, and temperature. After sonication, analytical size exclusion chromatography was used according to Example 1 conditions to confirm the relative size distributions of products. While absolute size and molecular weight determinations are unknown for the RCA products by this method, we observed the expected general trend of increasing elution time for smaller fragments. The following elution times were thus observed: 10.1 min (corresponding to the void volume) for ˜16 kb (theoretical maximum length) full length RCA product, 10.8 min for the 1.5 kb sonicated RCA product, 14.8 min for the 0.15 kb sonicated RCA product, 19.2 min for the starting 43b RCA template (control injection), 20.0 min for the unconjugated o25b(+)mrsa (control injection), and 23.7 min for residual small molecules including nucleotides.
  • FIGS. 10A and 10B shows that the larger the RCA product, the higher the activation (FIG. 10A % CD25 expression) and fold-expansion for the early days 1-4 of the experiment, while the trend becomes less pronounced later in the study. In particular, by day 7, all the three RCA products of different sizes yield comparable levels of cell expansion (FIG. 10B).
  • While only certain features of the invention have been illustrated and described herein, many modifications and changes will occur to those skilled in the art. It is, therefore, to be understood that the appended embodiments are intended to cover all such modifications and changes as fall within the scope of the invention.

Claims (25)

1. A method of activating T cells, the method comprising:
a) providing a population of T cells;
b) adding a plurality of first agents, wherein the first agent comprises a T-cell activator and a first binder moiety;
c) adding a second agent comprising a plurality of capture oligomers, wherein at least a segment of at least one of the plurality of capture oligomers is capable of associating with the first binder moiety; and
d) incubating the population of T cells after steps (b) and (c), whereby at least a portion of the population of T cells is activated.
2. The method of claim 1, wherein the second agent is a nucleic acid polymer comprising a plurality of capture oligonucleotide sequences.
3. The method of claim 2, wherein the second agent is a rolling circle amplification product comprising a plurality of capture oligonucleotide sequences.
4. The method of claim 2, wherein the first binder moiety is a nucleic acid sequence, wherein at least a segment of at least one of the plurality of capture oligonucleotide sequences is complementary to the nucleic acid sequence.
5. The method of claim 2, further comprising adding a nuclease.
6. The method of claim 1, wherein the T-cell activator is an anti-CD3 antibody or a fragment thereof.
7. The method of claim 1, further comprising adding a T-cell co-stimulator.
8. The method of claim 7 wherein the T-cell co-stimulator is an anti-CD28 antibody, an anti-CD2 antibody or fragments thereof.
9. The method of claim 7, wherein the T-cell co-stimulator is attached to a second binder moiety, wherein at least a segment of at least one of the plurality of capture oligomers is capable of associating with the second binder moiety.
10. The method of claim 9, wherein the second binder moiety is a nucleic acid sequence.
11. The method according to claim 7 where the T-cell activator is an anti-CD3 antibody attached to a nucleic acid sequence, the T-cell co-stimulator is an anti-CD28 antibody attached to a nucleic acid sequence, and the second agent is a rolling circle amplification product comprising of a plurality of complementary capture oligonucleotide sequences.
12. A method according to claim 1, further comprising a step of adding a vector comprising a foreign gene into the population of T cells.
13. A method according to claim 12, wherein the vector is added with the plurality of first agents.
14. A method according to claim 12, wherein the vector is added after at least a portion of the population of T cells is activated.
15. A method according to claim 12, wherein the vector is a lentivirus vector or a gamma-retrovirus vector.
16. A method of activating T cells, the method comprising:
a) providing a population of T cells;
b) adding a plurality of first agents, wherein the first agent comprises an anti-CD3 antibody attached to a nucleic acid sequence;
c) adding a nucleic acid polymer comprising a plurality of capture oligonucleotide sequences, wherein at least a segment of at least one of the plurality of capture oligonucleotide sequences is complementary to the nucleic acid sequence attached to the anti-CD3 antibody; and
d) incubating the population of T cells after steps (b) and (c), whereby at least a portion of the population of T cells is activated.
17. The method of claim 16, further comprising adding an anti-CD28 antibody.
18. The method of claim 17, wherein the anti-CD28 antibody is attached to a nucleic acid sequence, wherein at least a segment of at least one of the plurality of capture oligonucleotide sequences is complementary to the nucleic acid sequence attached to the anti-CD28 antibody.
19. A kit comprising:
a T-cell activator attached to a nucleic acid sequence;
a nucleic acid polymer comprising a plurality of capture oligonucleotide sequences, and
wherein at least a segment of at least one of the plurality of capture oligonucleotide sequences is complementary to the nucleic acid sequence attached to the T-cell activator.
20. The kit of claim 19, wherein the nucleic acid polymer is a rolling circle amplification product.
21. The kit of claim 19, wherein the T cell activator is an anti-CD3 antibody.
22. The kit of claim 19, further comprising a T-cell co-stimulator.
23. The kit if claim 22, wherein the T-cell co-stimulator is an anti-CD28 antibody.
24. The kit of claim 23, wherein the anti-CD28 antibody is attached to a nucleic acid sequence, wherein at least a segment of at least one of the plurality of capture oligonucleotide sequences is complementary to the nucleic acid sequence attached to the anti-CD28 antibody.
25. The kit of claim 19 further comprising a cell culture medium.
US15/245,584 2016-08-24 2016-08-24 Methods and kits for cell activation Active 2036-09-10 US10294454B2 (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
US15/245,584 US10294454B2 (en) 2016-08-24 2016-08-24 Methods and kits for cell activation
US16/339,077 US11512288B2 (en) 2016-08-24 2017-08-24 Methods and kits for cell activation
EP17769141.7A EP3504323A1 (en) 2016-08-24 2017-08-24 Methods and kits for cell activation
PCT/US2017/048306 WO2018039400A1 (en) 2016-08-24 2017-08-24 Methods and kits for cell activation
CN201780051673.1A CN109563484A (en) 2016-08-24 2017-08-24 Method and kit for cell activation
JP2019510693A JP7043114B2 (en) 2016-08-24 2017-08-24 Methods and kits for activating cells

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
US15/245,584 US10294454B2 (en) 2016-08-24 2016-08-24 Methods and kits for cell activation

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/339,077 Continuation US11512288B2 (en) 2016-08-24 2017-08-24 Methods and kits for cell activation

Publications (2)

Publication Number Publication Date
US20180057791A1 true US20180057791A1 (en) 2018-03-01
US10294454B2 US10294454B2 (en) 2019-05-21

Family

ID=59914514

Family Applications (2)

Application Number Title Priority Date Filing Date
US15/245,584 Active 2036-09-10 US10294454B2 (en) 2016-08-24 2016-08-24 Methods and kits for cell activation
US16/339,077 Active 2038-12-07 US11512288B2 (en) 2016-08-24 2017-08-24 Methods and kits for cell activation

Family Applications After (1)

Application Number Title Priority Date Filing Date
US16/339,077 Active 2038-12-07 US11512288B2 (en) 2016-08-24 2017-08-24 Methods and kits for cell activation

Country Status (5)

Country Link
US (2) US10294454B2 (en)
EP (1) EP3504323A1 (en)
JP (1) JP7043114B2 (en)
CN (1) CN109563484A (en)
WO (1) WO2018039400A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11512288B2 (en) 2016-08-24 2022-11-29 Global Life Sciences Solutions Usa Llc Methods and kits for cell activation

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB201820444D0 (en) * 2018-12-14 2019-01-30 Adaptimmune Ltd Marker for T cell expansion
CN110184239B (en) * 2019-06-12 2021-04-06 蓝莲(杭州)生物科技有限公司 Activated T lymphocyte culture solution and preparation method of activated T lymphocyte

Family Cites Families (105)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE3825615A1 (en) 1988-07-28 1990-02-01 Behringwerke Ag ANTIGENT CONSTRUCTS OF "MAJOR HISTOCOMPATIBILITY COMPLEX" CLASS I ANTIGENS WITH SPECIFIC CARRIER MOLECULES, THEIR PRODUCTION AND USE
US5858358A (en) 1992-04-07 1999-01-12 The United States Of America As Represented By The Secretary Of The Navy Methods for selectively stimulating proliferation of T cells
US6905680B2 (en) 1988-11-23 2005-06-14 Genetics Institute, Inc. Methods of treating HIV infected subjects
US6534055B1 (en) 1988-11-23 2003-03-18 Genetics Institute, Inc. Methods for selectively stimulating proliferation of T cells
US7479269B2 (en) * 1988-11-23 2009-01-20 Genetics Institute, Llc Methods for selectively enriching TH1 and TH2 cells
US6352694B1 (en) 1994-06-03 2002-03-05 Genetics Institute, Inc. Methods for inducing a population of T cells to proliferate using agents which recognize TCR/CD3 and ligands which stimulate an accessory molecule on the surface of the T cells
JP2618497B2 (en) 1989-10-03 1997-06-11 鐘淵化学工業株式会社 Tumor-damaging cell inducer and tumor-damaging cell induction device
US6197298B1 (en) 1991-04-19 2001-03-06 Tanox, Inc. Modified binding molecules specific for T lymphocytes and their use as in vivo immune modulators in animals
US6106835A (en) 1991-04-19 2000-08-22 Tanox, Inc. Modified binding molecules specific for T or B lymphocytes and their use as in vivo immune modulators
CA2065658A1 (en) 1991-04-19 1992-10-20 Tse-Wen Chang Conjugates of liposomes or microbeads and antibodies specific for t lymphocytes and their use in vivo immune modulators
US5872222A (en) 1991-04-19 1999-02-16 Tanox Biosystems, Inc. Conjugates of polymers and antibodies specific for T lymphocytes, and their use as adjuvants
US6129916A (en) 1991-04-19 2000-10-10 Tanox, Inc. Method of Increasing activation on proliferation of T cells using antibody-microbead conjugates
DE4237113B4 (en) 1992-11-03 2006-10-12 "Iba Gmbh" Peptides and their fusion proteins, expression vector and method of producing a fusion protein
WO1994012196A1 (en) 1992-11-25 1994-06-09 Tanox Biosystems, Inc. Conjugates and constructs including anti-cd28 and anti-cd3 binding molecules
EP0678034B1 (en) 1993-01-11 1999-05-26 Dana Farber Cancer Institute Inducing cytotoxic t lymphocyte responses
DK0700430T3 (en) 1993-06-04 2005-08-15 Us Navy Methods to selectively stimulate proliferation of T cells
CA2164646A1 (en) 1993-06-11 1994-12-22 Wade E. Bolton Anti-cd3 antibody-aminodextran conjugates for induction of t-cell activation and proliferation
US5635602A (en) 1993-08-13 1997-06-03 The Regents Of The University Of California Design and synthesis of bispecific DNA-antibody conjugates
US7175843B2 (en) 1994-06-03 2007-02-13 Genetics Institute, Llc Methods for selectively stimulating proliferation of T cells
EP0764203A1 (en) 1994-06-03 1997-03-26 THE UNITED STATES OF AMERICA as represented by THE SECRETARY OF THE NAVY Methods for selectively stimulating proliferation of t cells
US7074904B2 (en) 1994-07-29 2006-07-11 Altor Bioscience Corporation MHC complexes and uses thereof
CA2214649C (en) 1995-03-08 2007-06-12 Zeling Cai Antigen presenting system and methods for activation of t-cells
US7067318B2 (en) 1995-06-07 2006-06-27 The Regents Of The University Of Michigan Methods for transfecting T cells
JP4031033B2 (en) 1995-05-04 2008-01-09 アメリカ合衆国 Improved T cell transfection method
US6692964B1 (en) 1995-05-04 2004-02-17 The United States Of America As Represented By The Secretary Of The Navy Methods for transfecting T cells
NZ331688A (en) 1996-03-28 2000-02-28 Univ Johns Hopkins Soluble divalent and multivalent heterodimeric analogs of proteins
US7973137B1 (en) 1996-03-28 2011-07-05 Johns Hopkins University Cell compositions comprising molecular complexes that modify immune responses
US6458354B1 (en) 1996-03-28 2002-10-01 The Johns Hopkins University Molecular complexes which modify immune responses
ES2277358T3 (en) 1996-09-06 2007-07-01 Ortho-Mcneil Pharmaceutical, Inc. PURIFICATION OF ANTIGEN T SPECIFIC CELLS.
DE19641876B4 (en) 1996-10-10 2011-09-29 Iba Gmbh streptavidin muteins
US6077833A (en) 1996-12-31 2000-06-20 Isis Pharmaceuticals, Inc. Oligonucleotide compositions and methods for the modulation of the expression of B7 protein
US7927595B1 (en) 1997-02-21 2011-04-19 The United States Of America As Represented By The Secretary Of The Navy Methods for downregulating CCR5 in T cells with anti-CD3 antibodies and anti-CD28 antibodies
US6268411B1 (en) 1997-09-11 2001-07-31 The Johns Hopkins University Use of multivalent chimeric peptide-loaded, MHC/ig molecules to detect, activate or suppress antigen-specific T cell-dependent immune responses
FR2766205B1 (en) 1997-07-16 2002-08-30 Inst Nat Sante Rech Med NOVEL METHOD FOR SENSITIZING ANTIGEN PRESENTING CELLS AND NOVEL MEANS FOR IMPLEMENTING THE METHOD
US6248564B1 (en) 1997-08-29 2001-06-19 Harvard University Mutant MHC class I molecules
AU741130B2 (en) 1997-09-16 2001-11-22 Oregon Health Sciences University Recombinant MHC molecules useful for manipulation of antigen-specific T-cells
US6232445B1 (en) 1997-10-29 2001-05-15 Sunol Molecular Corporation Soluble MHC complexes and methods of use thereof
ES2279600T3 (en) 1998-03-12 2007-08-16 Miltenyi Biotec Gmbh MICROCOLUMN SYSTEM FOR MAGNETIC SEPARATION.
US20020051783A1 (en) 1998-06-05 2002-05-02 Savage Philip Michael Method for producing or enhancing a T-cell response against a target cell using a complex comprising an HLA class I molecule and an attaching means
GB2339782A (en) 1998-06-05 2000-02-09 Philip Michael Savage Chimeric protein complexes comprising HLA class I antigens
US7521197B2 (en) 1998-06-05 2009-04-21 Alexis Biotech Limited Method for producing cytotoxic T-cells
US7264965B2 (en) 1998-06-05 2007-09-04 Alexis Biotech Limited Method for producing or enhancing a T-cell response against a target cell using a complex comprising an HLA class I molecule and an attaching means
US6787154B2 (en) 1998-10-20 2004-09-07 Salvatore Albani Artificial antigen presenting cells
DK1123086T3 (en) 1998-10-20 2010-06-14 Androclus Technologies S R L I Artificial antigen-specific cells and associated methods
US6590076B1 (en) 1999-04-02 2003-07-08 Corixa Corporation Compositions for the treatment and diagnosis of breast cancer and methods for their use
US7541184B2 (en) 2000-02-24 2009-06-02 Invitrogen Corporation Activation and expansion of cells
US6867041B2 (en) 2000-02-24 2005-03-15 Xcyte Therapies, Inc. Simultaneous stimulation and concentration of cells
MXPA02008265A (en) 2000-02-24 2004-09-10 Xcyte Therapies Inc Simultaneous stimulation and concentration of cells.
US20030235908A1 (en) 2000-02-24 2003-12-25 Xcyte Therapies, Inc. Activation and expansion of cells
US20030119185A1 (en) 2000-02-24 2003-06-26 Xcyte Therapies, Inc. Activation and expansion of cells
US6797514B2 (en) 2000-02-24 2004-09-28 Xcyte Therapies, Inc. Simultaneous stimulation and concentration of cells
EP1526171A1 (en) 2000-02-24 2005-04-27 Xcyte Therapies, Inc Simultaneous stimulation and concentration of cells
US7572631B2 (en) 2000-02-24 2009-08-11 Invitrogen Corporation Activation and expansion of T cells
EP1330516B1 (en) 2000-11-03 2007-01-17 The Regents Of The University Of Michigan Surface transfection and expression procedure
US6902933B2 (en) 2000-11-03 2005-06-07 Regents Of The University Of Michigan Surface transfection and expression procedure
US6897067B2 (en) 2000-11-03 2005-05-24 Regents Of The University Of Michigan Surface transfection and expression procedure
EP1227321A1 (en) 2000-12-28 2002-07-31 Institut für Bioanalytik GmbH Reversible MHC multimer staining for functional purification of antigen-specific T cells
DE10113776B4 (en) 2001-03-21 2012-08-09 "Iba Gmbh" Isolated streptavidin-binding, competitively elutable peptide, this comprehensive fusion peptide, nucleic acid coding therefor, expression vector, methods for producing a recombinant fusion protein and methods for detecting and / or obtaining the fusion protein
US7745140B2 (en) 2002-01-03 2010-06-29 The Trustees Of The University Of Pennsylvania Activation and expansion of T-cells using an engineered multivalent signaling platform as a research tool
AU2003202908A1 (en) 2002-01-03 2003-07-24 The Trustees Of The University Of Pennsylvania Activation and expansion of t-cells using an engineered multivalent signaling platform
AU2003216436A1 (en) 2002-02-08 2003-09-02 Life Technologies Corporation Compositions and methods for restoring immune responsiveness in patients with immunological defects
CN100379852C (en) * 2002-02-08 2008-04-09 埃克斯西特治疗公司 Compositions and methods for restoring immune responsiveness in patients with immunological defects
US20040175373A1 (en) 2002-06-28 2004-09-09 Xcyte Therapies, Inc. Compositions and methods for eliminating undesired subpopulations of T cells in patients with immunological defects related to autoimmunity and organ or hematopoietic stem cell transplantation
US20050084967A1 (en) 2002-06-28 2005-04-21 Xcyte Therapies, Inc. Compositions and methods for eliminating undesired subpopulations of T cells in patients with immunological defects related to autoimmunity and organ or hematopoietic stem cell transplantation
WO2004003142A2 (en) 2002-06-28 2004-01-08 Xcyte Therapies, Inc. Compositions and methods for restoring immune repertoire in patients with immunological defects related to autoimmunity and organ or hematopoietic stem cell transplantation
JP5068931B2 (en) 2002-07-12 2012-11-07 ザ ジョンズ ホプキンス ユニバーシティー Reagents and methods for binding to lymphocyte receptors of unique clonal traits
GB2392158B (en) 2002-08-21 2005-02-16 Proimmune Ltd Chimeric MHC protein and oligomer thereof
CA2525519A1 (en) 2003-05-08 2004-12-02 Xcyte Therapies, Inc. Generation and isolation of antigen-specific t cells
US7402431B2 (en) 2004-03-01 2008-07-22 Immunovative Therapies, Ltd. T-cell therapy formulation
US7435592B2 (en) 2003-05-13 2008-10-14 Immunovative Therapies, Ltd. Compositions for allogeneic cell therapy
WO2005049085A1 (en) 2003-11-18 2005-06-02 Valeocyte Therapies Llc Use of soluble complexes to facilitate cell activation
US20050175583A1 (en) 2003-12-02 2005-08-11 Lawrence Tamarkin Methods and compositions for the production of monoclonal antibodies
US7592431B2 (en) 2004-02-26 2009-09-22 Immunovative Therapies, Ltd. Biodegradable T-cell Activation device
US7678572B2 (en) 2004-02-26 2010-03-16 Immunovative Therapies, Ltd. Methods for preparing T-cells for cell therapy
DE502004010393D1 (en) 2004-03-04 2009-12-31 Leukocare Gmbh Leukocyte stimulation matrix
GB2422834B8 (en) 2005-02-04 2007-01-04 Proimmune Ltd MHC oligomer and method of making the same
JP2006345852A (en) * 2005-06-16 2006-12-28 Virxsys Corp Antibody conjugate
GB0603081D0 (en) 2006-02-15 2006-03-29 Dynal Biotech Asa Oslo Method
GB2442048B (en) 2006-07-25 2009-09-30 Proimmune Ltd Biotinylated MHC complexes and their uses
CA2665568C (en) 2006-10-04 2018-01-09 Janssen Pharmaceutica, N.V. Preparation of inactivated artificial antigen presenting cells and their use in cell therapies
WO2008116468A2 (en) 2007-03-26 2008-10-02 Dako Denmark A/S Mhc peptide complexes and uses thereof in infectious diseases
WO2009005552A2 (en) 2007-03-29 2009-01-08 Epitype Corporation Methods and compositions for multivalent binding and methods for manufacture of rapid diagnostic tests
US8629098B2 (en) 2008-01-15 2014-01-14 Yale University Compositions and methods for adoptive and active immunotherapy
US8658178B2 (en) 2008-03-19 2014-02-25 Yale University Carbon nanotube compositions and methods of use thereof
WO2010037395A2 (en) 2008-10-01 2010-04-08 Dako Denmark A/S Mhc multimers in cancer vaccines and immune monitoring
WO2011050000A2 (en) 2009-10-20 2011-04-28 The Regents Of The University Of California Single molecule nucleic acid nanoparticles
GB201002730D0 (en) 2010-02-18 2010-04-07 Uni I Oslo Product
WO2012024695A1 (en) 2010-08-20 2012-02-23 Life Technologies Corporation Magnetic beads having surface glycoconjugates and use thereof
WO2012044999A2 (en) 2010-10-01 2012-04-05 Ludwig Institute For Cancer Research Ltd. Reversible protein multimers, methods for their production and use
CN103797028B (en) 2011-07-18 2017-08-25 Iba 股份有限公司 Make the method for the reversible dyeing of target cell
US8834889B2 (en) 2011-08-29 2014-09-16 National Tsing Hua University Antigen presenting composition and use thereof
JP2015500296A (en) 2011-12-09 2015-01-05 ザ・ジョンズ・ホプキンス・ユニバーシティ Artificial antigen-presenting cells with a defined dynamic shape
WO2013109568A1 (en) 2012-01-17 2013-07-25 Croda, Inc. Seal swell additive
CN106940268B (en) 2012-02-23 2021-09-21 朱诺治疗有限公司 Chromatographic separation of cells and other complex biological materials
EP2711418B1 (en) * 2012-09-25 2017-08-23 Miltenyi Biotec GmbH Method for polyclonal stimulation of T cells by flexible nanomatrices
WO2014076277A1 (en) 2012-11-16 2014-05-22 Iba Gmbh Streptavidin muteins and methods of using them
ES2820729T3 (en) 2013-02-15 2021-04-22 Univ Johns Hopkins Antigen-specific T-cell redirectors
JP6501270B2 (en) 2013-03-14 2019-04-17 ザ カリフォルニア インスティテュート フォー バイオメディカル リサーチ Targeting agent-antibody conjugates and uses thereof
CN105431523B (en) 2013-03-14 2020-08-21 约翰·霍普金斯大学 Nanoscale artificial antigen presenting cells
BR112015032277B1 (en) 2013-06-24 2020-12-01 Neximmune antigen presentation complex, its use, and composition
ES2612918T3 (en) * 2013-07-10 2017-05-19 Miltenyi Biotec Gmbh Method to induce the proliferation of natural killer cells by mobile nanomatrices
JP5799058B2 (en) 2013-07-30 2015-10-21 三桜工業株式会社 Negative pressure pump and cylinder head cover
CN116656605A (en) 2014-04-16 2023-08-29 朱诺治疗有限公司 Methods, kits and devices for expanding cell populations
WO2016033690A1 (en) * 2014-09-04 2016-03-10 Stemcell Technologies Inc. Soluble antibody complexes for t cell or nk cell activation and expansion
US10294454B2 (en) 2016-08-24 2019-05-21 General Electric Company Methods and kits for cell activation

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11512288B2 (en) 2016-08-24 2022-11-29 Global Life Sciences Solutions Usa Llc Methods and kits for cell activation

Also Published As

Publication number Publication date
JP2019534684A (en) 2019-12-05
EP3504323A1 (en) 2019-07-03
CN109563484A (en) 2019-04-02
WO2018039400A1 (en) 2018-03-01
US10294454B2 (en) 2019-05-21
US20200181572A1 (en) 2020-06-11
JP7043114B2 (en) 2022-03-29
US11512288B2 (en) 2022-11-29

Similar Documents

Publication Publication Date Title
US20240084287A1 (en) Single cell cellular component enrichment from barcoded sequencing libraries
AU2020230334A1 (en) Compositions and methods for immunooncology
CA3126699A1 (en) Modified immune cells having enhanced anti-neoplasia activity and immunosuppression resistance
JP2018512162A5 (en)
CA3151690A1 (en) Genetically-edited immune cells and methods of therapy
JP2004024264A (en) ANTISENSE-OLIGONUCLEOTIDES FOR TREATMENT OF IMMUNOSUPPRESSIVE EFFECTS OF TRANSFORMING GROWTH FACTOR-beta (TGF-beta)
US10294454B2 (en) Methods and kits for cell activation
Cardle et al. Biomaterials in chimeric antigen receptor T-cell process development
CA3168932A1 (en) Methods for activation and expansion of tumor infiltrating lymphocytes
JP2021518760A (en) Modification of loci in the immune-related genome using paired CRISPR nickaseribonuclear proteins
US20240024478A1 (en) Compositions and Methods for Reducing HLA-A in a Cell
US20210062248A1 (en) Methods of performing guide-seq on primary human t cells
IL303505A (en) Compositions and methods for reducing mhc class ii in a cell
CA3183129A1 (en) Rna scaffolds
US20240139323A1 (en) Compositions and Methods for Genetically Modifying CIITA in a Cell
US20210171908A1 (en) Allogeneic t-cells and methods for production thereof
RU2771624C2 (en) Compositions and methods for immune oncology
WO2023240218A1 (en) Methods for detecting genomic abnormalities in cells
KR20230044537A (en) Compositions and methods for engineering and selecting CAR T cells with desired phenotypes

Legal Events

Date Code Title Description
AS Assignment

Owner name: GENERAL ELECTRIC COMPANY, NEW YORK

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:KOVACS, ERNEST WILLIAM;SOOD, ANUP;SMITH, REGINALD DONOVAN;AND OTHERS;SIGNING DATES FROM 20160729 TO 20160824;REEL/FRAME:039525/0388

STPP Information on status: patent application and granting procedure in general

Free format text: NOTICE OF ALLOWANCE MAILED -- APPLICATION RECEIVED IN OFFICE OF PUBLICATIONS

AS Assignment

Owner name: GENERAL ELECTRIC COMPANY, NEW YORK

Free format text: CORRECTIVE ASSIGNMENT TO CORRECT THE FIFTH ASSIGNOR CHADHURI, PADMAPARNA PREVIOUSLY RECORDED ON REEL 039525 FRAME 0388. ASSIGNOR(S) HEREBY CONFIRMS THE ASSIGNMENT;ASSIGNORS:KOVACS, ERNEST WILLIAM;SOOD, ANUP;SMITH, REGINALD DONOVAN;AND OTHERS;SIGNING DATES FROM 20160729 TO 20190320;REEL/FRAME:050719/0263

STPP Information on status: patent application and granting procedure in general

Free format text: PUBLICATIONS -- ISSUE FEE PAYMENT VERIFIED

STCF Information on status: patent grant

Free format text: PATENTED CASE

AS Assignment

Owner name: GLOBAL LIFE SCIENCES SOLUTIONS USA LLC, MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:GENERAL ELECTRIC COMPANY;REEL/FRAME:053966/0133

Effective date: 20200320

MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 4TH YEAR, LARGE ENTITY (ORIGINAL EVENT CODE: M1551); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

Year of fee payment: 4